165 results on '"Seong Who Kim"'
Search Results
2. Concomitant induction of SLIT3 and microRNA-218–2 in macrophages by toll-like receptor 4 activation limits osteoclast commitment
- Author
-
Eun-Young Kim, Ji-Eun Kim, Soo-Hyun Chung, Ji-Eun Park, Dohee Yoon, Hyo-Jin Min, Yoolim Sung, Soo Been Lee, Seong Who Kim, and Eun-Ju Chang
- Subjects
SLIT3 ,Macrophages ,Toll-like receptor 4 (TLR4) ,miR-218–2-3p ,RANK ,Osteoclast commitment ,Medicine ,Cytology ,QH573-671 - Abstract
Abstract Background Toll-like receptor 4 (TLR4) conducts a highly regulated inflammatory process by limiting the extent of inflammation to avoid toxicity and tissue damage, even in bone tissues. Thus, it is plausible that strategies for the maintenance of normal bone-immunity to prevent undesirable bone damage by TLR4 activation can exist, but direct evidence is still lacking. Methods Osteoclast precursors (OCPs) obtained from WT or Slit3-deficient mice were differentiated into osteoclast (OC) with macrophage colony-stimulating factor (M-CSF), RANK ligand (RANKL) and lipopolysaccharide (LPS) by determining the number of TRAP-positive multinuclear cells (TRAP+ MNCs). To determine the alteration of OCPs population, fluorescence-activated cell sorting (FACS) was conducted in bone marrow cells in mice after LPS injection. The severity of bone loss in LPS injected WT or Slit3-deficient mice was evaluated by micro-CT analysis. Result We demonstrate that TLR4 activation by LPS inhibits OC commitment by inducing the concomitant expression of miR-218–2-3p and its host gene, Slit3, in mouse OCPs. TLR4 activation by LPS induced SLIT3 and its receptor ROBO1 in BMMs, and this SLIT3-ROBO1 axis hinders RANKL-induced OC differentiation by switching the protein levels of C/EBP-β isoforms. A deficiency of SLIT3 resulted in increased RANKL-induced OC differentiation, and the elevated expression of OC marker genes including Pu.1, Nfatc1, and Ctsk. Notably, Slit3-deficient mice showed expanded OCP populations in the bone marrow. We also found that miR-218–2 was concomitantly induced with SLIT3 expression after LPS treatment, and that this miRNA directly suppressed Tnfrsf11a (RANK) expression at both gene and protein levels, linking it to a decrease in OC differentiation. An endogenous miR-218–2 block rescued the expression of RANK and subsequent OC formation in LPS-stimulated OCPs. Aligned with these results, SLIT3-deficient mice displayed increased OC formation and reduced bone density after LPS challenge. Conclusion Our findings suggest that the TLR4-dependent concomitant induction of Slit3 and miR-218–2 targets RANK in OCPs to restrain OC commitment, thereby avoiding an uncoordinated loss of bone through inflammatory processes. These observations provide a mechanistic explanation for the role of TLR4 in controlling the commitment phase of OC differentiation. Video Abstract
- Published
- 2023
- Full Text
- View/download PDF
3. miR-3189-targeted GLUT3 repression by HDAC2 knockdown inhibits glioblastoma tumorigenesis through regulating glucose metabolism and proliferation
- Author
-
Sungmin Kwak, Seung-Ho Park, Sung-Hak Kim, Gi-Jun Sung, Ji-Hye Song, Ji-Hoon Jeong, Hyunhee Kim, Chang Hoon Ha, Seong Who Kim, and Kyung-Chul Choi
- Subjects
HDAC2 ,Glioblastoma ,Glioma stem cells ,miR-3189 ,GLUT3 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Epigenetic regulations frequently appear in Glioblastoma (GBM) and are highly associated with metabolic alterations. Especially, Histone deacetylases (HDACs) correlates with the regulation of tumorigenesis and cell metabolism in GBM progression, and HDAC inhibitors report to have therapeutic efficacy in GBM and other neurological diseases; however, GBM prevention and therapy by HDAC inhibition lacks a mechanism in the focus of metabolic reprogramming. Methods HDAC2 highly express in GBM and is analyzed in TCGA/GEPIA databases. Therefore, HDAC2 knockdown affects GBM cell death. Analysis of RNA sequencing and qRT-PCR reveals that miR-3189 increases and GLUT3 decreases by HDAC2 knockdown. GBM tumorigenesis also examines by using in vivo orthotopic xenograft tumor models. The metabolism change in HDAC2 knockdown GBM cells measures by glucose uptake, lactate production, and OCR/ECAR analysis, indicating that HDAC2 knockdown induces GBM cell death by inhibiting GLUT3. Results Notably, GLUT3 was suppressed by increasing miR-3189, demonstrating that miR-3189-mediated GLUT3 inhibition shows an anti-tumorigenic effect and cell death by regulating glucose metabolism in HDAC2 knockdown GBM. Conclusions Our findings will demonstrate the central role of HDAC2 in GBM tumorigenesis through the reprogramming of glucose metabolism by controlling miR-3189-inhibited GLUT3 expression, providing a potential new therapeutic strategy for GBM treatment.
- Published
- 2022
- Full Text
- View/download PDF
4. Heterologous ChAdOx1-BNT162b2 vaccination in Korean cohort induces robust immune and antibody responses that includes Omicron
- Author
-
Hye Kyung Lee, Jinyoung Go, Heungsup Sung, Seong Who Kim, Mary Walter, Ludwig Knabl, Priscilla A. Furth, Lothar Hennighausen, and Jin Won Huh
- Subjects
Health sciences ,Immunology ,Immune response ,Science - Abstract
Summary: Heterologous ChAdOx1-BNT162b2 vaccination induces a stronger immune response than BNT162b2-BNT162b2. Here, we investigated the molecular transcriptome, germline allelic variants of immunoglobulin loci, and anti-Omicron antibody levels in 46 office and lab workers from the Republic of Korea following ChAdOx1-BNT162b2 vaccination. Anti-spike-specific IgG antibody levels against the ancestral SARS-CoV-2 strain increased from 70 AU/ml to 14,000 AU/ml to 142,000 AU/ml one, three and seven days following the second vaccination. Titers against VOC, including Omicron, were two-fold to three-fold lower, yet higher than those measured following BNT162b2-BNT162b2 vaccination. RNA-seq of peripheral immune cells demonstrated activation of interferon pathways with increased IGHV clonal transcripts encoding neutralizing antibodies. scRNA-seq revealed enriched B cell and CD4+ T cell responses in both ChAdOx1-BNT162b2 and BNT162b2-BNT162b2 recipients, but a stronger clonal expansion of memory B cells with ChAdOx1-BNT162b2. In summary, heterologous ChAdOx1-BNT162b2 provides an innate and adaptive immune response that exceeds homologous BNT162b2 vaccination.
- Published
- 2022
- Full Text
- View/download PDF
5. miR-351-5p/Miro2 axis contributes to hippocampal neural progenitor cell death via unbalanced mitochondrial fission
- Author
-
Ha-Na Woo, Sujeong Park, Hae Lin Kim, Min-Kyo Jung, Chan-Gi Pack, Jinsu Park, Yoonsuk Cho, Dong-Gyu Jo, Dong Kyu Kim, Inhee Mook-Jung, Seong Who Kim, and Heuiran Lee
- Subjects
hippocampal neural progenitor cells ,miRNA-351-5p ,Miro GTPase ,cell death ,autophagy ,mitophagy ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Adult hippocampal neurogenesis supports the structural and functional plasticity of the brain, while its decline is associated with neurodegeneration common in Alzheimer’s disease (AD). Although the dysregulation of certain microRNAs (miRNAs) in AD have been observed, the effects of miRNAs on hippocampal neurogenesis are largely unknown.In this study, we demonstrated miR-351-5p as a causative factor in hippocampal neural progenitor cell death through modulation of the mitochondrial guanosine triphosphatase (GTPase), Miro2. Downregulation of Miro2 by siMiro2 induced cell death, similar to miR-351-5p, whereas ectopic Miro2 expression using an adenovirus abolished these effects. Excessively fragmented mitochondria and dysfunctional mitochondria were indexed by decreased mitochondrial potential, and increased reactive oxygen species were identified in miR-351-5p-induced cell death. Moreover, subsequent induction of mitophagy via Pink1 and Parkin was observed in the presence of miR-351-5p and siMiro2. The suppression of mitochondrial fission by Mdivi-1 completely inhibited cell death by miR-351-5p. miR-351-5p expression increased whereas the level of Miro2 decreased in the hippocampus of AD model mice, emulating expression in AD patients. Collectively, the data indicate the mitochondrial fission and accompanying mitophagy by miR-351-5p/Miro2 axis as critical in hippocampal neural progenitor cell death, and a potential therapeutic target in AD.
- Published
- 2021
- Full Text
- View/download PDF
6. Epigenetic regulation of p62/SQSTM1 overcomes the radioresistance of head and neck cancer cells via autophagy-dependent senescence induction
- Author
-
Myungjin Lee, Hae Yun Nam, Hee-Bum Kang, Won Hyeok Lee, Geun-Hee Lee, Gi-Jun Sung, Myung Woul Han, Kyung-Ja Cho, Eun-Ju Chang, Kyung-Chul Choi, Seong Who Kim, and Sang Yoon Kim
- Subjects
Cytology ,QH573-671 - Abstract
Abstract Tumors are composed of subpopulations of cancer cells with functionally distinct features. Intratumoral heterogeneity limits the therapeutic effectiveness of cancer drugs. To address this issue, it is important to understand the regulatory mechanisms driving a subclonal variety within a therapy-resistant tumor. We identified tumor subclones of HN9 head and neck cancer cells showing distinct responses to radiation with different levels of p62 expression. Genetically identical grounds but epigenetic heterogeneity of the p62 promoter regions revealed that radioresistant HN9-R clones displayed low p62 expression via the creation of repressive chromatin architecture, in which cooperation between DNMT1 (DNA methyltransferases 1) and HDAC1 (histone deacetylases 1) resulted in DNA methylation and repressive H3K9me3 and H3K27me3 marks in the p62 promoter. Combined inhibition of DNMT1 and HDAC1 by genetic depletion or inhibitors enhanced the suppressive effects on proliferative capacity and in vivo tumorigenesis following irradiation. Importantly, ectopically p62-overexpressed HN9-R clones increased the induction of senescence along with p62-dependent autophagy activation. These results demonstrate the heterogeneous expression of p62 as the key component of clonal variation within a tumor against irradiation. Understanding the epigenetic diversity of p62 heterogeneity among subclones allows for improved identification of the functional state of subclones and provides a novel treatment option to resolve resistance to current therapies.
- Published
- 2021
- Full Text
- View/download PDF
7. CD26 Inhibition Potentiates the Therapeutic Effects of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells by Delaying Cellular Senescence
- Author
-
Miyeon Kim, Jinyoung Go, Ji Hye Kwon, Hye Jin Jin, Yun Kyung Bae, Eun-Young Kim, Eun-Ju Chang, Soo Jin Choi, and Seong Who Kim
- Subjects
CD26 ,DPP4 ,mesenchymal stem cell ,cellular senescence ,emphysema ,Biology (General) ,QH301-705.5 - Abstract
Mesenchymal stem cells (MSCs) are recognized as potential treatments for multiple degenerative and inflammatory disorders as a number of animal and human studies have indicated their therapeutic effects. There are also several clinically approved medicinal products that are manufactured using these cells. For such large-scale manufacturing requirements, the in vitro expansion of harvested MSCs is essential. Multiple subculturing of MSCs, however, provokes cellular senescence processes which is known to deteriorate the therapeutic efficacy of the cells. Strategies to rejuvenate or selectively remove senescent MSCs are therefore highly desirable for fostering future clinical applications of these cells. In this present study, we investigated gene expression changes related to cellular senescence of MSCs derived from umbilical cord blood and found that CD26, also known as DPP4, is significantly upregulated upon cellular aging. We further observed that the inhibition of CD26 by genetic or pharmacologic means delayed the cellular aging of MSCs with their multiple passaging in culture. Moreover, the sorting and exclusion of CD26-positive MSCs from heterogenous cell population enhanced in vitro cell attachment and reduced senescence-associated cytokine secretion. CD26-negative MSCs also showed superior therapeutic efficacy in mouse lung emphysema model. Our present results collectively suggest CD26 is a potential novel target for the rejuvenation of senescent MSCs for their use in manufacturing MSC-based applications.
- Published
- 2022
- Full Text
- View/download PDF
8. Mesenchymal stem cells prevent the progression of diabetic nephropathy by improving mitochondrial function in tubular epithelial cells
- Author
-
Seung Eun Lee, Jung Eun Jang, Hyun Sik Kim, Min Kyo Jung, Myoung Seok Ko, Mi-Ok Kim, Hye Sun Park, Wonil Oh, Soo Jin Choi, Hye Jin Jin, Sang-Yeob Kim, Yun Jae Kim, Seong Who Kim, Min Kyung Kim, Chang Ohk Sung, Chan-Gi Pack, Ki-Up Lee, and Eun Hee Koh
- Subjects
Medicine ,Biochemistry ,QD415-436 - Abstract
Kidney disease: stem cells to the rescue Stem cells can halt the progression of kidney damage owing to diabetes by reducing inflammation and improving energy production in kidney cells. Eun Hee Koh at the University of Ulsan College of Medicine in Seoul, South Korea, and colleagues found that adult stem cells, known as mesenchymal stem cells (MSCs), derived from human umbilical cord blood had a protective effect on the kidneys of diabetic mice. Repeated administration of MSCs prevented the recruitment of pro-inflammatory cells into the kidney and increased the levels of arginase-1, a marker of cells with anti-inflammatory activity. Experiments in cells showed that MSCs stimulated the production of arginase-1 in that, in turn, were able to increase the production and activity of mitochondria in kidney cells. This study confirms an important role for MSCs in organ repair.
- Published
- 2019
- Full Text
- View/download PDF
9. The Upregulation of Toll-Like Receptor 3 via Autocrine IFN-β Signaling Drives the Senescence of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells Through JAK1
- Author
-
Hyang Ju Lee, Bongkun Choi, Yongsub Kim, Sang Eun Lee, Hye Jin Jin, Hee-Seop Lee, Eun-Ju Chang, and Seong Who Kim
- Subjects
senescence ,mesenchymal stromal cell (MSC) ,Toll-like receptor 3 (TLR3) ,Janus kinase 1 (JAK1) ,interferon-β (IFN-β) ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Although mesenchymal stromal cells (MSCs) are among the most promising cell sources for cell-based therapies and regenerative medicine, the decline in their function with age due to cellular senescence limits their therapeutic applications. Unveiling the underlying mechanism of MSC senescence is therefore of substantial interest with regard to advancing MSC-based cell therapies. We here show that the induction of human umbilical cord blood-derived MSC (UCB-MSC) senescence causes the predominant upregulation of Toll-like receptor 3 (TLR3). Subsequent TLR3 activation by polyinosinic-polycytidylic acid triggers the prominent features of senescence. Using a clustered regularly interspaced short palindromic repeats/Cas9 library screening system, we identified Janus kinase 1 (JAK1) as the candidate regulatory factor for TLR3-mediated MSC senescence. A JAK1 deficiency blocked the MSC senescence phenotype upon TLR3 activation and TLR3 induction. Targeting the JAK1 pathway using chemical JAK1 inhibitors also significantly suppressed TLR3-mediated MSC senescence. Importantly, we further observed that UCB-MSC senescence is driven by a senescence-associated secretory phenotype (SASP) and that interferon-β (IFN-β) is a component of TLR3-dependent SASP, whereby its autocrine actions upregulate TLR3 and suppress cell proliferation. A JAK1 depletion significantly interrupted these effects of IFN-β, indicating that JAK1 is a signaling mediator linking IFN-β activity to TLR3 expression and the process of MSC senescence. Collectively, our findings provide new mechanistic insights into UCB-MSC senescence by revealing the role of an autocrine regulatory loop of SASP evoked by TLR3 activation.
- Published
- 2019
- Full Text
- View/download PDF
10. Elevated Neuropeptide Y in Endothelial Dysfunction Promotes Macrophage Infiltration and Smooth Muscle Foam Cell Formation
- Author
-
Bongkun Choi, Min-Kyung Shin, Eun-Young Kim, Ji-Eun Park, Halim Lee, Seong Who Kim, Jae-Kwan Song, and Eun-Ju Chang
- Subjects
neuropeptide Y (NPY) ,pentraxin 3 (PTX3) ,smooth muscle foam cell ,macrophage ,lipid ,endothelial nitric oxide synthase (eNOS) ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Endothelial dysfunction has been linked to vascular inflammation and foam cell formation but the underlying mechanisms still remain unclear. We sought to define the factors inducing inflammation and smooth muscle foam cell formation under endothelial dysfunction using endothelial nitric oxide synthase (eNOS)-deficient mice. Vascular smooth muscle cells (VSMCs) from eNOS-deficient mice displayed increased expression of macrophage-related genes and elevated lipid uptake. Neuropeptide Y (NPY) was upregulated in the aorta from the eNOS-deficient mice and promoted macrophage chemotaxis toward VSMCs while enhancing the activity of matrix metalloproteinase-3. Notably, NPY induced lipid uptake in VSMCs, facilitating smooth muscle foam cell formation, in association with enhanced expression of genes related to modified low-density lipoprotein uptake and macrophages. NPY was augmented by inflammatory pentraxin 3 (PTX3) in VSMCs. PTX3 enhanced macrophage migratory capacity through the NPY/neuropeptide Y receptor axis and this effect was attenuated by pharmacological inhibition with a receptor-specific antagonist. These observations suggest that endothelial dysfunction leads to the elevation of NPY that amplifies vascular inflammation by increasing inflammatory cell chemotaxis and triggers smooth muscle foam cell formation.
- Published
- 2019
- Full Text
- View/download PDF
11. Progressive Impairment of NK Cell Cytotoxic Degranulation Is Associated With TGF-β1 Deregulation and Disease Progression in Pancreatic Cancer
- Author
-
Eunsung Jun, Ah Young Song, Ji-Wan Choi, Hyeon Ho Lee, Mi-Yeon Kim, Dae-Hyun Ko, Hyo Jeong Kang, Seong Who Kim, Yenan Bryceson, Song Cheol Kim, and Hun Sik Kim
- Subjects
natural killer cells ,pancreatic cancer ,cytotoxicity ,TGF-β1 ,prognosis ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Natural killer (NK) cells are key effectors in cancer immunosurveillance and can be used as a prognostic biomarker in diverse cancers. Nonetheless, the role of NK cells in pancreatic cancer (PC) remains elusive, given conflicting data on their association with disease prognosis. In this study, using conventional K562 target cells and complementary engineered target cells providing defined and synergistic stimulation for NK cell activation, a correlation between impaired NK cell cytotoxic degranulation and PC progression was determined. Peripheral blood mononuclear cells (PBMCs) from 31 patients with newly diagnosed PC, 24 patients with non-malignant tumors, and 37 healthy controls were analyzed by flow cytometry. The frequency, phenotype, and effector functions of the NK cells were evaluated, and correlations between NK cell functions and disease stage and prognosis were analyzed. The results demonstrated that effector functions, but not frequency, of NK cells was progressively decreased on a per-cell basis during PC progression. Impaired cytotoxic degranulation, but not IFN-γ production, was associated with clinical features indicating disease progression, such as high serum CA19-9 and high-grade tumors. Significantly, this impairment correlated with cancer recurrence and mortality in a prospective analysis. Furthermore, the impaired cytotoxic degranulation was unrelated to NKG2D downregulation but was associated with increased circulating and tumor-associated TGF-β1 expression. Thus, NK cell cytotoxic activity was associated with PC progression and may be a favorable biomarker with predictive and prognostic value in PC.
- Published
- 2019
- Full Text
- View/download PDF
12. Tristetraprolin Posttranscriptionally Downregulates TRAIL Death Receptors
- Author
-
Won Hyeok Lee, Myung Woul Han, Song Hee Kim, Daseul Seong, Jae Hee An, Hyo Won Chang, Sang Yoon Kim, Seong Who Kim, and Jong Cheol Lee
- Subjects
tumor necrosis factor-related apoptosis-inducing ligand ,death receptor ,tristetraprolin ,posttranscriptional modification ,AU-rich elements ,cancer treatment ,Cytology ,QH573-671 - Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has attracted attention as a potential candidate for cancer therapy. However, many primary cancers are resistant to TRAIL, even when combined with standard chemotherapy. The mechanism of TRAIL resistance in cancer cells has not been fully elucidated. The TRAIL death receptor (DR) 3′-untranslated region (3′-UTR) is reported to contain AU-rich elements (AREs) that are important for regulating DR mRNA stability. However, the mechanisms by which DR mRNA stability is determined by its 3′-UTR are unknown. We demonstrate that tristetraprolin (TTP), an ARE-binding protein, has a critical function of regulating DR mRNA stability. DR4 mRNA contains three AREs and DR5 mRNA contains four AREs in 3′-UTR. TTP bound to all three AREs in DR4 and ARE3 in DR5 and enhanced decay of DR4/5 mRNA. TTP overexpression in colon cancer cells changed the TRAIL-sensitive cancer cells to TRAIL-resistant cells, and down-regulation of TTP increased TRAIL sensitivity via DR4/5 expression. Therefore, this study provides a molecular mechanism for enhanced levels of TRAIL DRs in cancer cells and a biological basis for posttranscriptional modification of TRAIL DRs. In addition, TTP status might be a biomarker for predicting TRAIL response when a TRAIL-based treatment is used for cancer.
- Published
- 2020
- Full Text
- View/download PDF
13. Elevated Pentraxin 3 in Obese Adipose Tissue Promotes Adipogenic Differentiation by Activating Neuropeptide Y Signaling
- Author
-
Min-Kyung Shin, Bongkun Choi, Eun-Young Kim, Ji-Eun Park, Eui Seung Hwang, Hyang Ju Lee, Min Kyung Kim, Ji-Eun Kim, Seong Who Kim, and Eun-Ju Chang
- Subjects
pentraxin3 ,neuropeptide Y ,obesity ,adipogenesis ,macrophage ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Obesity is accompanied by chronic systemic inflammation characterized by macrophage infiltration of obese tissues, an elevated plasma level of inflammatory substances, and excessive accumulation of lipids. The pro-inflammatory factor pentraxin 3 (PTX3) is also elevated in obese tissues, suggesting its potential role in adipogenesis. We found by analyzing murine preadipocyte 3T3-L1 cells, and human adipocytes derived from mesenchymal stem cells, which locally elevated PTX3 in obese adipose tissue augments adipocyte differentiation and subsequent lipid accumulation. This occurs via the upregulation of adipogenesis-related transcription factors. PTX3 enhanced lipid accumulation in murine 3T3-L1 cells by upregulating the expression of neuropeptide Y (NPY)/NPY receptor (NPYR) expression in preadipocytes. Pharmacological inhibition by NPYR antagonists abolished these effects. NPY also promoted the production of reactive oxygen species (ROS), a known trigger of adipogenesis. NPYR antagonists as well as antioxidant N-acetylcysteine showed anti-adipogenic effects by reducing the ROS levels, indicating that PTX3 mediates adipogenesis through NPY-dependent ROS production. These findings suggest that PTX3 plays a key role in the development of obesity by enhancing adipocyte differentiation and lipid synthesis via NPY/NPYR signaling. These observations provide a mechanistic explanation for the adipogenesis mediated by PTX3.
- Published
- 2018
- Full Text
- View/download PDF
14. Degeneration of the nigrostriatal pathway and induction of motor deficit by tetrahydrobiopterin: an in vivo model relevant to Parkinson’s disease
- Author
-
Seong Who Kim, Yeon Joo Jang, Jin Woo Chang, and Onyou Hwang
- Subjects
Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
We determined whether the preferential toxicity of tetrahydrobiopterin (BH4) on dopamine-producing cells, which we have previously observed in vitro, might also occur in vivo and generate characteristics associated with Parkinson’s disease. Intrastriatal BH4 injection caused a loss of tyrosine hydroxylase immunoreactivity and decreased dopamine content. The dopaminergic cell bodies topologically corresponding to the lesioned terminals were selectively degenerated. This was accompanied by a dose-dependent and asymmetric movement deficit in the contralateral forepaw. Direct injection of BH4 into the substantia nigra caused a loss of tyrosine hydroxylase immunoreactivity, but injection into the dorsal raphe was without effect on the GTP cyclohydrolase-immunoreactive serotonergic neurons, demonstrating selectivity for the dopaminergic system. BH4 exhibited a range of potency comparable to that of 6-hydroxydopamine. Thus, this animal model generated by the administration of BH4, the molecule endogenously present in the monoaminergic neurons, exhibited morphological, biochemical, and behavioral characteristics associated with Parkinson’s disease and may be useful for studies in dopaminergic degeneration.
- Published
- 2003
- Full Text
- View/download PDF
15. Letter-to-editor, 'Feedback amplification of senolysis using caspase-cleavable peptide-doxorubicin conjugate and 2DG' [Journal of Controlled Release, Volume 346, pp. 158-168, (2022), doi: 10.1016/j.jconrel.2022.04.012]
- Author
-
Jong Cheol Lee, Gui Chul Kim, Seong Who Kim, Na Kyeong Lee, Young Seok Cho, Seung Woo Chung, Yoon Se Lee, Myung Woul Han, Hyo Won Chang, Youngro Byun, and Sang Yoon Kim
- Subjects
Pharmaceutical Science - Published
- 2023
16. Supplementary Figure Legends from Radioresistant Cancer Cells Can Be Conditioned to Enter Senescence by mTOR Inhibition
- Author
-
Seong Who Kim, Sang Yoon Kim, Neung Hwa Park, Seung-Ho Choi, Hyesung Jeon, Min Kyo Jung, Kyung Eun Lee, Hee Jin Lee, Byoung Wook Lee, Hyang Ju Lee, Myungjin Lee, Yoon Sun Lee, Hyo Won Chang, Myung Woul Han, and Hae Yun Nam
- Abstract
Supplementary Figure Legends - PDF file 151K, Legend for Supplementary Figures S1-S6
- Published
- 2023
17. Supplementary Figure S2 from Radioresistant Cancer Cells Can Be Conditioned to Enter Senescence by mTOR Inhibition
- Author
-
Seong Who Kim, Sang Yoon Kim, Neung Hwa Park, Seung-Ho Choi, Hyesung Jeon, Min Kyo Jung, Kyung Eun Lee, Hee Jin Lee, Byoung Wook Lee, Hyang Ju Lee, Myungjin Lee, Yoon Sun Lee, Hyo Won Chang, Myung Woul Han, and Hae Yun Nam
- Abstract
Supplementary Figure S2 PDF - PDF file 414K, Activation of autophagy in AMC-HN-9 cells
- Published
- 2023
18. Supplementary Methods from Radioresistant Cancer Cells Can Be Conditioned to Enter Senescence by mTOR Inhibition
- Author
-
Seong Who Kim, Sang Yoon Kim, Neung Hwa Park, Seung-Ho Choi, Hyesung Jeon, Min Kyo Jung, Kyung Eun Lee, Hee Jin Lee, Byoung Wook Lee, Hyang Ju Lee, Myungjin Lee, Yoon Sun Lee, Hyo Won Chang, Myung Woul Han, and Hae Yun Nam
- Abstract
Supplementary Methods - PDF file 168K, Additional experimental procedures, including western blot, RT-PCR, cell cycle analysis, etc. Also includes supplementary references
- Published
- 2023
19. ZBTB7A suppresses glioblastoma tumorigenesis through the transcriptional repression of EPB41L5
- Author
-
Ji-Hoon Jeong, Seung-Ho Park, Hyunhee Kim, Hae Yun Nam, Sung-Hak Kim, Minseok Jeong, Min-Jeong Kong, Jihyun Son, Ji-Eun Jeong, Ji-Hye Song, Seong Who Kim, and Kyung-Chul Choi
- Subjects
Clinical Biochemistry ,Molecular Medicine ,Molecular Biology ,Biochemistry - Abstract
Glioblastoma multiforme (GBM), the most aggressive and malignant glioma, has poor prognosis. Although patients with GBM are treated with surgery, chemotherapy, and radiation therapy, GBM is highly resistant to treatment, difficult to treat, and expensive. In this study, we analyzed the Gene Expression Profiling Interactive Analysis dataset, the Cancer Genome Atlas dataset, and Gene Expression Omnibus array data. ZBTB7A (also called FBI1/POKEMON/LRF) was found to be highly expressed in low-grade GBM, but significantly reduced in patients with GBM. ZBTB7A is a transcription factor that plays an important role in many developmental stages, including cell proliferation. The activation of epithelial-mesenchymal transition (EMT) in cancer is a key process in cancer progression and metastasis. Erythrocyte membrane protein band 4.1 Like 5 (EPB41L5) is an essential protein for EMT progression and metastasis in various types of cancer. We found that ZBTB7A-depleted U87 cells exhibited GBM progression and metastasis. Based on RNA sequencing data, ZBTB7A directly binds to the promoter of the EPB41L5 gene, reducing its expression and inhibiting GBM progression. We demonstrated that ZBTB7A dramatically inhibited GBM tumor growth through transcriptional repression of EPB41L5. Thus, both ZBTB7A and EPB41L5 may be potential biomarkers and novel therapeutic targets for GBM treatment. Taken together, we discovered the role of a novel tumor suppressor, ZBTB7A, that directly inhibits GBM progression, and EPB41L5 is a therapeutic target protein for patients with GBM and an essential protein for the development of GBM therapeutics.
- Published
- 2022
20. Tristetraprolin regulates phagocytosis through interaction with CD47 in head and neck cancer
- Author
-
Won Hyeok, Lee, Song Hee, Kim, Jae Hee, An, Tae-Koon, Kim, Hee Jeong, Cha, Hyo Won, Chang, Sang Yoon, Kim, Seong Who, Kim, and Myung Woul, Han
- Subjects
Cancer Research ,Immunology and Microbiology (miscellaneous) ,General Medicine - Abstract
CD47 is expressed in all human cancer cells, including head and neck cancer, and initiates a signaling cascade to inhibit macrophage phagocytosis. However, the mechanism underlying CD47 overexpression has not been elucidated in radioresistant head and neck cancer. The present study demonstrated that decreased Tristetraprolin (TTP) expression induced a sustained overexpression of CD47 using reverse transcription-quantitative PCR and western blotting, and that CD47 overexpression prevented phagocytosis using a phagocytosis assay in a radioresistant HN31R cell line. Subsequently, using TTP transfection, RNA interference, duel-luciferase assay and EMSA, it was revealed that TTP transfection enhanced phagocytosis through degradation of CD47 mRNA by directly binding to CD47 AREs within the CD47 3'UTR. Based on our previous study, methylation-specific PCR and western blotting revealed that DNMT1 was overexpressed in radioresistant HN31R cell line and TTP expression was decreased epigenetically by DMNT1 associated DNA methylation. Overall, these findings provided novel insight into the role of TTP as a biomarker of CD47-positive head and neck cancer patients.
- Published
- 2022
21. Optimal Ratio of Wnt3a Expression in Human Mesenchymal Stem Cells Promotes Axonal Regeneration in Spinal Cord Injured Rat Model
- Author
-
Jeong Hoon Kim, Hyung Ho Yoon, Heuiran Lee, Seong Who Kim, Sang Ryong Jeon, Hyang Ju Lee, Jin Hoon Park, Joongkee Min, and Ji Hyun Kim
- Subjects
animal structures ,030218 nuclear medicine & medical imaging ,Andrology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Spinal cord injuries ,Gap-43 protein ,Spinal cord injury ,Spinal Cord Regeneration ,biology ,business.industry ,General Neuroscience ,Regeneration (biology) ,Recovery of function ,Mesenchymal stem cell ,Wnt3a ,medicine.disease ,Spine ,Transplantation ,Spinal cord regeneration ,embryonic structures ,biology.protein ,Laboratory Investigation ,Mesenchymal stem cells ,Surgery ,Neurology (clinical) ,Stem cell ,business ,030217 neurology & neurosurgery ,Immunostaining - Abstract
Objective : Through our previous clinical trials, the demonstrated therapeutic effects of MSC in chronic spinal cord injury (SCI) were found to be not sufficient. Therefore, the need to develop stem cell agent with enhanced efficacy is increased. We transplanted enhanced Wnt3asecreting human mesenchymal stem cells (hMSC) into injured spines at 6 weeks after SCI to improve axonal regeneration in a rat model of chronic SCI. We hypothesized that enhanced Wnt3a protein expression could augment neuro-regeneration after SCI.Methods : Thirty-six Sprague-Dawley rats were injured using an Infinite Horizon (IH) impactor at the T9–10 vertebrae and separated into five groups : 1) phosphate-buffered saline injection (injury only group, n=7); 2) hMSC transplantation (MSC, n=7); 3) hMSC transfected with pLenti vector (without Wnt3a gene) transplantation (pLenti-MSC, n=7); 4) hMSC transfected with Wnt3a gene transplantation (Wnt3a-MSC, n=7); and 5) hMSC transfected with enhanced Wnt3a gene (1.7 fold Wnt3a mRNA expression) transplantation (1.7 Wnt3a-MSC, n=8). Six weeks after SCI, each 5×105 cells/15 µL at 2 points were injected using stereotactic and microsyringe pump. To evaluate functional recovery from SCI, rats underwent Basso-Beattie-Bresnahan (BBB) locomotor test on the first, second, and third days post-injury and then weekly for 14 weeks. Axonal regeneration was assessed using growth-associated protein 43 (GAP43), microtubule-associated protein 2 (MAP2), and neurofilament (NF) immunostaining.Results : Fourteen weeks after injury (8 weeks after transplantation), BBB score of the 1.7 Wnt3a-MSC group (15.0±0.28) was significantly higher than that of the injury only (10.0±0.48), MSC (12.57±0.48), pLenti-MSC (12.42±0.48), and Wnt3a-MSC (13.71±0.61) groups (p
- Published
- 2021
22. MDM2-dependent Sirt1 degradation is a prerequisite for Sirt6-mediated cell death in head and neck cancers
- Author
-
So Young Cheon, Jong Sil Lee, Ji Hyun Seo, Young-Sool Hah, Sang Yoon Kim, Hyo Won Chang, Jung Je Park, Jeong Seok Hwa, Seong Jun Won, Somi Ryu, and Seong Who Kim
- Subjects
SIRT6 ,Male ,Programmed cell death ,Clinical Biochemistry ,Cell ,Mice, Nude ,Apoptosis ,medicine.disease_cause ,Biochemistry ,Article ,Mice ,Downregulation and upregulation ,Sirtuin 1 ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Medicine ,Animals ,Humans ,Sirtuins ,Molecular Biology ,Cell Proliferation ,chemistry.chemical_classification ,Reactive oxygen species ,biology ,business.industry ,Oral cancer ,Proto-Oncogene Proteins c-mdm2 ,Prognosis ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Survival Rate ,enzymes and coenzymes (carbohydrates) ,medicine.anatomical_structure ,chemistry ,Head and Neck Neoplasms ,Sirtuin ,Proteolysis ,biology.protein ,Cancer research ,Molecular Medicine ,Mdm2 ,business ,Carcinogenesis ,Reactive Oxygen Species ,hormones, hormone substitutes, and hormone antagonists - Abstract
Sirt6 is involved in multiple biological processes, including aging, metabolism, and tumor suppression. Sirt1, another member of the sirtuin family, functionally overlaps with Sirt6, but its role in tumorigenesis is controversial. In this study, we focused on cell death in association with Sirt6/Sirt1 and reactive oxygen species (ROS) in head and neck squamous cell carcinomas (HNSCCs). Sirt6 induced cell death, as widely reported, but Sirt1 contributed to cell death only when it was suppressed by Sirt6 via regulation of MDM2. Sirt6 and Sirt6-mediated suppression of Sirt1 upregulated ROS, which further led to HNSCC cell death. These results provide insight into the molecular roles of Sirt6 and Sirt1 in tumorigenesis and could therefore contribute to the development of novel strategies to treat HNSCC., Cancer: A trio of proteins to tackle tumors New understanding of the interactions between three proteins sheds light on their role in either promoting or restricting the development of tumors called squamous cell carcinomas, which account for over 90% of all cancers in the head and neck. Researchers in South Korea led by Sang Yoon Kim and Seong Who Kim at the University of Ulsan, Seoul, investigated the role of the proteins Sirt6, Sirt1 and MDM2 in controlling the death of cancer cells caused by chemicals called reactive oxygen species (ROS). The effects of Sirt6 and Sirt1 combine to regulate ROS-induced cancer cell death. Sirt6 controls the activity of MDM2, stimulating ROS production. Sirt6 also influences MDM2 to suppress Sirt1 activity, thereby also promoting cancer cell death. Drugs affecting these three proteins could offer new approaches to anti-cancer therapy.
- Published
- 2021
23. miR-351-5p/Miro2 axis contributes to hippocampal neural progenitor cell death via unbalanced mitochondrial fission
- Author
-
Jinsu Park, Chan-Gi Pack, S. S. Park, Ha Na Woo, Min Kyo Jung, Dong Kyu Kim, Yoonsuk Cho, Inhee Mook-Jung, Dong-Gyu Jo, Seong Who Kim, Hae Lin Kim, and Heuiran Lee
- Subjects
0301 basic medicine ,autophagy ,PINK1 ,Mitochondrion ,Hippocampal formation ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,Mitophagy ,miRNA-351-5p ,medicine ,Progenitor cell ,Miro GTPase ,hippocampal neural progenitor cells ,lcsh:RM1-950 ,Neurogenesis ,Neurodegeneration ,medicine.disease ,Cell biology ,lcsh:Therapeutics. Pharmacology ,cell death ,mitophagy ,030104 developmental biology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Original Article ,Mitochondrial fission ,Alzheimer’s disease - Abstract
Adult hippocampal neurogenesis supports the structural and functional plasticity of the brain, while its decline is associated with neurodegeneration common in Alzheimer’s disease (AD). Although the dysregulation of certain microRNAs (miRNAs) in AD have been observed, the effects of miRNAs on hippocampal neurogenesis are largely unknown. In this study, we demonstrated miR-351-5p as a causative factor in hippocampal neural progenitor cell death through modulation of the mitochondrial guanosine triphosphatase (GTPase), Miro2. Downregulation of Miro2 by siMiro2 induced cell death, similar to miR-351-5p, whereas ectopic Miro2 expression using an adenovirus abolished these effects. Excessively fragmented mitochondria and dysfunctional mitochondria were indexed by decreased mitochondrial potential, and increased reactive oxygen species were identified in miR-351-5p-induced cell death. Moreover, subsequent induction of mitophagy via Pink1 and Parkin was observed in the presence of miR-351-5p and siMiro2. The suppression of mitochondrial fission by Mdivi-1 completely inhibited cell death by miR-351-5p. miR-351-5p expression increased whereas the level of Miro2 decreased in the hippocampus of AD model mice, emulating expression in AD patients. Collectively, the data indicate the mitochondrial fission and accompanying mitophagy by miR-351-5p/Miro2 axis as critical in hippocampal neural progenitor cell death, and a potential therapeutic target in AD., Graphical Abstract, Woo et al. revealed that miR-351-5p is the causative factor for hippocampal neural progenitor cell death through modulation of Miro2. They suggest that mitochondrial fission by the miR-351-5p/Miro2 axis, and accompanying mitophagy, is a critical event in hippocampal neural progenitor cell death, which could be a potential therapeutic target in AD.
- Published
- 2021
24. Genetic immune response and antibody repertoire of heterologous ChAdOx1-BNT162b2 vaccination in a Korean cohort
- Author
-
Hye Kyung Lee, Jinyoung Go, Heungsup Sung, Seong Who Kim, Mary Walter, Ludwig Knabl, Priscilla Furth, Lothar Hennighausen, and Jin Won Huh
- Abstract
Heterologous ChAdOx1-BNT162b2 vaccination induces a stronger immune response than two doses of BNT162b2 or ChAdOx1. Yet, the molecular transcriptome, the germline allelic variants of immunoglobulin loci and anti-Omicron antibody levels induced by the heterologous vaccination have not been formally investigated. Moreover, there is a paucity of COVID vaccine studies including diverse genetic populations. Here, we show a robust molecular immune transcriptome and antibody repertoire in 51 office workers from the Republic of Korea after a heterologous ChAdOx1-BNT162b2 vaccination or a homologous ChAdOx1-ChAdOx1 vaccination. Anti-spike-specific IgG antibody levels in the heterologous group increased from 14,000 U/ml to 142,000 AU/ml within eight days after the BNT162b2 vaccination. In contrast, antibody levels in the homologous group increased two-fold after the second ChAdOx1 dose. Antibody titers against the Omicron spike protein as compared to the ancestral strain were reduced to a lesser extent in the heterologous group. RNA-seq conducted on immune cells demonstrated a stronger activation of interferon-induced genetic programs in the heterologous cohort. An increase of specific IGHV clonal transcripts encoding neutralizing antibodies was preferentially detected in the heterologous cohort. Enrichment of B cell and CD4+ T cell responses were observed following both heterologous and homologous vaccination using scRNA-seq, but clonally expanded memory B cells were relatively stronger in the ChAdOx1-BNT162b2 cohort. In summary, a heterologous vaccination with ChAdOx1 followed by BNT162b2 provides an innate and adaptive immune response exceeding that seen in homologous ChAdOx1 vaccinations but equivalent to that seen in homologous BNT162b2 vaccination.
- Published
- 2022
25. miR-3189-targeted GLUT3 repression by HDAC2 knockdown inhibits glioblastoma tumorigenesis through regulating glucose metabolism and proliferation
- Author
-
Sungmin Kwak, Seung-Ho Park, Sung-Hak Kim, Gi-Jun Sung, Ji-Hye Song, Ji-Hoon Jeong, Hyunhee Kim, Chang Hoon Ha, Seong Who Kim, and Kyung-Chul Choi
- Subjects
Gene Expression Regulation, Neoplastic ,Cancer Research ,MicroRNAs ,Glucose ,Oncology ,Glucose Transporter Type 3 ,Brain Neoplasms ,Carcinogenesis ,Cell Line, Tumor ,Histone Deacetylase 2 ,Humans ,Glioblastoma ,Cell Proliferation - Abstract
Background Epigenetic regulations frequently appear in Glioblastoma (GBM) and are highly associated with metabolic alterations. Especially, Histone deacetylases (HDACs) correlates with the regulation of tumorigenesis and cell metabolism in GBM progression, and HDAC inhibitors report to have therapeutic efficacy in GBM and other neurological diseases; however, GBM prevention and therapy by HDAC inhibition lacks a mechanism in the focus of metabolic reprogramming. Methods HDAC2 highly express in GBM and is analyzed in TCGA/GEPIA databases. Therefore, HDAC2 knockdown affects GBM cell death. Analysis of RNA sequencing and qRT-PCR reveals that miR-3189 increases and GLUT3 decreases by HDAC2 knockdown. GBM tumorigenesis also examines by using in vivo orthotopic xenograft tumor models. The metabolism change in HDAC2 knockdown GBM cells measures by glucose uptake, lactate production, and OCR/ECAR analysis, indicating that HDAC2 knockdown induces GBM cell death by inhibiting GLUT3. Results Notably, GLUT3 was suppressed by increasing miR-3189, demonstrating that miR-3189-mediated GLUT3 inhibition shows an anti-tumorigenic effect and cell death by regulating glucose metabolism in HDAC2 knockdown GBM. Conclusions Our findings will demonstrate the central role of HDAC2 in GBM tumorigenesis through the reprogramming of glucose metabolism by controlling miR-3189-inhibited GLUT3 expression, providing a potential new therapeutic strategy for GBM treatment.
- Published
- 2021
26. Impaired migration of autologous induced neural stem cells from patients with schizophrenia and implications for genetic risk for psychosis
- Author
-
Junhee Lee, Sehyeon Song, Juhee Lee, Jisoo Kang, Eun Kyung Choe, Tae Young Lee, Myong-Wuk Chon, Minah Kim, Seong Who Kim, Myung-Suk Chun, Mi-Sook Chang, and Jun Soo Kwon
- Subjects
Psychiatry and Mental health ,Neural Stem Cells ,Psychotic Disorders ,Schizophrenia ,Humans ,Biological Psychiatry - Abstract
Stem cell technologies have presented explicit evidence of the neurodevelopmental hypothesis of schizophrenia. However, few studies investigated relevance of the schizophrenia genetic liability and the use of genetic reprogramming on pluripotent stem cells to the impaired neurodevelopment shown by stem cells. Therefore, this study sought to investigate the cellular phenotypes of induced neural stem cells (iNSCs) derived without genetic modification from patients with schizophrenia and from genetic high risk (GHR) individuals. Three patients with a diagnosis of schizophrenia, 3 GHR individuals who had two or more relatives with schizophrenia, and 3 healthy volunteers participated. iNSCs were derived using a small molecule-based lineage switch method, and their gene expression levels and migration capabilities were examined. Demographic characteristics were not different among the groups (age, χ
- Published
- 2021
27. CD26 Inhibition Potentiates the Therapeutic Effects of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells by Delaying Cellular Senescence
- Author
-
Miyeon Kim, Jinyoung Go, Ji Hye Kwon, Hye Jin Jin, Yun Kyung Bae, Eun-Young Kim, Eun-Ju Chang, Soo Jin Choi, and Seong Who Kim
- Subjects
emphysema ,QH301-705.5 ,cellular senescence ,Cell Biology ,DPP4 ,Biology (General) ,mesenchymal stem cell ,CD26 ,Developmental Biology - Abstract
Mesenchymal stem cells (MSCs) are recognized as potential treatments for multiple degenerative and inflammatory disorders as a number of animal and human studies have indicated their therapeutic effects. There are also several clinically approved medicinal products that are manufactured using these cells. For such large-scale manufacturing requirements, the in vitro expansion of harvested MSCs is essential. Multiple subculturing of MSCs, however, provokes cellular senescence processes which is known to deteriorate the therapeutic efficacy of the cells. Strategies to rejuvenate or selectively remove senescent MSCs are therefore highly desirable for fostering future clinical applications of these cells. In this present study, we investigated gene expression changes related to cellular senescence of MSCs derived from umbilical cord blood and found that CD26, also known as DPP4, is significantly upregulated upon cellular aging. We further observed that the inhibition of CD26 by genetic or pharmacologic means delayed the cellular aging of MSCs with their multiple passaging in culture. Moreover, the sorting and exclusion of CD26-positive MSCs from heterogenous cell population enhanced in vitro cell attachment and reduced senescence-associated cytokine secretion. CD26-negative MSCs also showed superior therapeutic efficacy in mouse lung emphysema model. Our present results collectively suggest CD26 is a potential novel target for the rejuvenation of senescent MSCs for their use in manufacturing MSC-based applications.
- Published
- 2021
28. Knockout of β2 Microglobulin Potentiates The Effects of IFN-γ Priming On The Survival and Immunomodulatory Capacities of Mesenchymal Stem Cells
- Author
-
Seong Who Kim, A.Reum Han, Sang Eun Lee, Soo Bin Lee, Jiyeon Kweon, Eun-Ju Chang, Jinyoung Go, Ha Rim Shin, and Yongsub Kim
- Subjects
Beta-2 microglobulin ,Chemistry ,Mesenchymal stem cell ,Cancer research ,Priming (immunology) - Abstract
Background MSCs have long been thought to be immune-privileged with low levels of major histocompatibility complex (MHC) class I and rare expression of MHC class II. However, growing evidence indicates that these cells may not actually be hypoimmunogenic, particularly when exposed to cytokines such as IFN-γ. IFN-γ primed increase of MHC class I expression can promote the rejection of allogenic MSCs in the host recipient. A strategy to overcome this drawback is urgently required. Methods We knocked out β2-microglobulin (B2M) in MSCs, which is a component of MHC class I, using the ribonucleoprotein (RNP)-mediated clustered regularly interspaced short palindromic repeats (CRISPR)- CRISPR-associated protein 9 (Cas9) system. The expression of MSC surface markers, MHC class I, and B2M was assayed by flow cytometry and western blotting. Upon co-culture of MSCs with CD4+ and CD8+ T cells, the survival and proliferation of both cell types were examined by cell counting kit (CCK-8) and carboxy fluorescein succinimidyl ester (CFSE), respectively. The levels of immunomodulatory molecules in MSCs were evaluated by both enzyme-linked immunosorbent assay (ELISA) and western blotting. Results B2M-knockout MSCs expressed low levels of MHC class I even upon IFN-γ priming, but maintained their native properties as evidenced by the expression of specific surface markers. CD8+ T cell proliferation was also far less stimulated by B2M-knockout MSCs than by control cells. Under these conditions, B2M-knockout MSCs had a significantly longer survival duration (> 2.4 fold) than did control cells. B2M-knockout MSCs showed significantly elevated levels of immune-modulatory molecules including indoleamine 2, 3-dioxygenase 1 (IDO-1), prostaglandin E2 (PGE2), C-C motif chemokine ligand 2 (CCL-2), and interleukin-6 (IL-6); conversely, B2M-knockout MSCs produced significantly lower levels of proinflammatory molecules (e.g., IL-1b, CXCL10) compared with control cells. Conclusion The loss of B2M in MSCs potentiated the immunomodulatory effects of IFN-γ priming while mitigating its potential inflammatory effects. B2M-knockout MSCs are a potentially promising treatment for immune-related inflammatory diseases.
- Published
- 2021
29. Branched-chain amino acids sustain pancreatic cancer growth by regulating lipid metabolism
- Author
-
Jaekyoung Son, Ji Hye Kim, Ji Hyeon Lee, Young-Ra Cho, Jong-Wook Kim, Seong Who Kim, and Hae Yun Nam
- Subjects
Clinical Biochemistry ,Glutamic Acid ,BCKDHA ,Mice, SCID ,Pregnancy Proteins ,Biochemistry ,Article ,Minor Histocompatibility Antigens ,Cell growth ,Adenosine Triphosphate ,Oxygen Consumption ,Pancreatic cancer ,medicine ,Animals ,Humans ,Metabolomics ,RNA, Messenger ,Molecular Biology ,Transaminases ,Cancer ,Cell Proliferation ,Gene knockdown ,Catabolism ,Chemistry ,Lentivirus ,Lipid metabolism ,Metabolism ,Lipid Metabolism ,medicine.disease ,Pancreatic Neoplasms ,Lipogenesis ,Cancer research ,Molecular Medicine ,Female ,Reactive Oxygen Species ,Amino Acids, Branched-Chain ,Carcinoma, Pancreatic Ductal - Abstract
Branched-chain amino acid (BCAA) catabolism and high levels of enzymes in the BCAA metabolic pathway have recently been shown to be associated with cancer growth and survival. However, the precise roles of BCAA metabolism in cancer growth and survival remain largely unclear. Here, we found that BCAA metabolism has an important role in human pancreatic ductal adenocarcinoma (PDAC) growth by regulating lipogenesis. Compared with nontransformed human pancreatic ductal (HPDE) cells, PDAC cells exhibited significantly elevated BCAA uptake through solute carrier transporters, which were highly upregulated in pancreatic tumor tissues compared with normal tissues. Branched-chain amino-acid transaminase 2 (BCAT2) knockdown markedly impaired PDAC cell proliferation, but not HPDE cell proliferation, without significant alterations in glutamate or reactive oxygen species levels. Furthermore, PDAC cell proliferation, but not HPDE cell proliferation, was substantially inhibited upon knockdown of branched-chain α-keto acid dehydrogenase a (BCKDHA). Interestingly, BCKDHA knockdown had no significant effect on mitochondrial metabolism; that is, neither the level of tricarboxylic acid cycle intermediates nor the oxygen consumption rate was affected. However, BCKDHA knockdown significantly inhibited fatty-acid synthesis, indicating that PDAC cells may utilize BCAAs as a carbon source for fatty-acid biosynthesis. Overall, our findings show that the BCAA metabolic pathway may provide a novel therapeutic target for pancreatic cancer., Pancreatic cancer: targeting a link between amino acids and lipids Essential nutrient molecules called branched-chain amino acids (BCAAs) sustain pancreatic cancer by supporting the formation of fatty ‘lipid’ compounds, offering possible new approaches for treatment. Jaekyoung Son and colleagues at the University of Ulsan College of Medicine in Seoul, South Korea, explored the role of BCAAs in the most common form of pancreatic cancer, pancreatic ductal adenocarcinoma. Effective treatments are urgently required as the disease is generally diagnosed too late for surgery, the most effective treatment, to be successful. The researchers found that human cancer cells exploit increased uptake of BCAAs as a source of carbon to build lipids. Disabling the genes for enzymes that convert BCAAs into lipids significantly and selectively reduced cancer cell growth. Drugs that interfere with key points in the over-active uptake of BCAA and conversion to lipids could be developed into new therapies.
- Published
- 2019
30. Links between accelerated replicative cellular senescence and down-regulation of SPHK1 transcription
- Author
-
Min Kyung Kim, Wooseong Lee, Gang-Ho Yoon, Eun-Ju Chang, Sun-Cheol Choi, and Seong Who Kim
- Subjects
Senescence ,Stromal cell ,Cell division ,Human adipose-derived stromal cells ,Down-Regulation ,Apoptosis ,Biochemistry ,Sphingolipid ,03 medical and health sciences ,chemistry.chemical_compound ,Downregulation and upregulation ,Sphingosine ,Humans ,Sphingosine kinase 1 ,SPHK1 transcription ,Molecular Biology ,Cellular Senescence ,Cell Proliferation ,0303 health sciences ,Gene knockdown ,Fumonisin B1 ,Sphingolipids ,biology ,Chemistry ,030302 biochemistry & molecular biology ,Mesenchymal Stem Cells ,General Medicine ,Articles ,Cell biology ,Phosphotransferases (Alcohol Group Acceptor) ,biology.protein ,Replicative senescence ,Lysophospholipids - Abstract
We have identified a mechanism to diminish the proliferative capacity of cells during cell expansion using human adiposederived stromal cells (hAD-SCs) as a model of replicative senescence. hAD-SCs of high-passage numbers exhibited a reduced proliferative capacity with accelerated cellular senescence. Levels of key bioactive sphingolipids were significantly increased in these senescent hAD-SCs. Notably, the transcription of sphingosine kinase 1 (SPHK1) was down-regulated in hAD-SCs at high-passage numbers. SPHK1 knockdown as well as inhibition of its enzymatic activity impeded the proliferation of hAD-SCs, with concomitant induction of cellular senescence and accumulation of sphingolipids, as seen in high-passage cells. SPHK1 knockdown-accelerated cellular senescence was attenuated by co-treatment with sphingosine-1-phosphate and an inhibitor of ceramide synthesis, fumonisin B1, but not by treatment with either one alone. Together, these results suggest that transcriptional down-regulation of SPHK1 is a critical inducer of altered sphingolipid profiles and enhances replicative senescence during multiple rounds of cell division. [BMB Reports 2019; 52(3): 220-225].
- Published
- 2019
31. Endothelial dysfunction induces atherosclerosis: increased aggrecan expression promotes apoptosis in vascular smooth muscle cells
- Author
-
Sang-Min Kim, Eun-Ju Chang, Ji Eun Park, Wooseong Lee, Jae-Wan Huh, Seong Who Kim, Eun Young Kim, Min Kyung Shin, and Bongkun Choi
- Subjects
Vascular smooth muscle ,Nitric Oxide Synthase Type III ,Apoptosis ,Biochemistry ,Muscle, Smooth, Vascular ,Mice ,03 medical and health sciences ,Enos ,Vascular smooth muscle cells ,Animals ,Humans ,Medicine ,Aggrecans ,Endothelial dysfunction ,Molecular Biology ,Cells, Cultured ,Aggrecan ,Cell Proliferation ,Mice, Knockout ,0303 health sciences ,biology ,business.industry ,Cell growth ,030302 biochemistry & molecular biology ,Endothelial Cells ,Articles ,General Medicine ,Atherosclerosis ,musculoskeletal system ,medicine.disease ,biology.organism_classification ,Plaque, Atherosclerotic ,Mice, Inbred C57BL ,Proteoglycan ,biology.protein ,Cancer research ,Endothelial nitric oxide synthase ,Signal transduction ,business ,Signal Transduction - Abstract
Endothelial dysfunction-induced lipid retention is an early feature of atherosclerotic lesion formation. Apoptosis of vascular smooth muscle cells (VSMCs) is one of the major modulating factors of atherogenesis, which accelerates atherosclerosis progression by causing plaque destabilization and rupture. However, the mechanism underlying VSMC apoptosis mediated by endothelial dysfunction in relation to atherosclerosis remains elusive. In this study, we reveal differential expression of several genes related to lipid retention and apoptosis, in conjunction with atherosclerosis, by utilizing a genetic mouse model of endothelial nitric oxide synthase (eNOS) deficiency manifesting endothelial dysfunction. Moreover, eNOS deficiency led to the enhanced susceptibility against pro-apoptotic insult in VSMCs. In particular, the expression of aggrecan, a major proteoglycan, was elevated in aortic tissue of eNOS deficient mice compared to wild type mice, and administration of aggrecan induced apoptosis in VSMCs. This suggests that eNOS deficiency may elevate aggrecan expression, which promotes apoptosis in VSMC, thereby contributing to atherosclerosis progression. These results may facilitate the development of novel approaches for improving the diagnosis or treatment of atherosclerosis. [BMB Reports 2019; 52(2): 145-150].
- Published
- 2019
32. p53/BNIP3-dependent mitophagy limits glycolytic shift in radioresistant cancer
- Author
-
Hye Min Lee, Kyung Eun Lee, Jong Cheol Lee, Sang Yoon Kim, Hyang Ju Lee, Seong Who Kim, Hyo Won Chang, Mi Ra Kim, Myungjin Lee, Hae Yun Nam, Gui Chul Kim, Youngro Byun, Yoon Sun Lee, and Hye Jin Jang
- Subjects
0301 basic medicine ,Cancer Research ,Cancer ,Biology ,Mitochondrion ,medicine.disease ,Head and neck squamous-cell carcinoma ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cell culture ,030220 oncology & carcinogenesis ,Radioresistance ,Mitophagy ,Cancer cell ,Genetics ,medicine ,Cancer research ,Glycolysis ,Molecular Biology - Abstract
The role of p53 in genotoxic therapy-induced metabolic shift in cancers is not yet known. In this study, we investigated the role of p53 in the glycolytic shift in head and neck squamous cell carcinoma cell lines following irradiation. Isogenic p53-null radioresistant cancer cells established through cumulative irradiation showed decreased oxygen consumption and increased glycolysis with compromised mitochondria, corresponding with their enhanced sensitivity to drugs that target glycolysis. In contrast, radioresistant cancer cells with wild-type p53 preserved their primary metabolic profile with intact mitophagic processes and maintained their mitochondrial integrity. Moreover, we identified a previously unappreciated link between p53 and mitophagy, which limited the glycolytic shift through the BNIP3-dependent clearance of abnormal mitochondria. Thus, drugs targeting glycolysis could be used as an alternative strategy for overcoming radioresistant cancers, and the p53 status could be used as a biomarker for selecting participants for clinical trials.
- Published
- 2019
33. Mitophagy deficiency increases NLRP3 to induce brown fat dysfunction in mice
- Author
-
Myoung Seok Ko, Sean M. Hartig, Seong Who Kim, In Jeoung Baek, David A. Bader, Ki Up Lee, Joong Yeol Park, Chul-Ho Lee, Eun Hee Koh, David D. Moore, Inkyu Lee, Jung Eun Jang, Seung Ho Heo, Ji Young Yun, Jaeseok Han, Jae Man Lee, Jung Jin Hwang, Eun Gyoung Hong, Seung Eun Lee, Jong Seok Moon, and Un Jung Kang
- Subjects
0301 basic medicine ,Biochemistry & Molecular Biology ,Inflammasomes ,white adipocyte ,Adipose tissue ,PINK1 ,Biology ,Brown adipocyte ,03 medical and health sciences ,Adipose Tissue, Brown ,inflammasome ,NLR Family, Pyrin Domain-Containing 3 Protein ,Mitophagy ,Brown adipose tissue ,Adipocytes ,Autophagy ,medicine ,Animals ,transcriptional activation ,pink1 ,Molecular Biology ,Gene knockout ,Mice, Knockout ,030102 biochemistry & molecular biology ,Inflammasome ,Cell Biology ,Thermogenin ,Mitochondria ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Biochemistry and Cell Biology ,Energy Metabolism ,Reactive Oxygen Species ,Research Article ,Research Paper ,medicine.drug - Abstract
Although macroautophagy/autophagy deficiency causes degenerative diseases, the deletion of essential autophagy genes in adipocytes paradoxically reduces body weight. Brown adipose tissue (BAT) plays an important role in body weight regulation and metabolic control. However, the key cellular mechanisms that maintain BAT function remain poorly understood. in this study, we showed that global or brown adipocyte-specific deletion of pink1, a Parkinson disease-related gene involved in selective mitochondrial autophagy (mitophagy), induced BAT dysfunction, and obesity-prone type in mice. Defective mitochondrial function is among the upstream signals that activate the NLRP3 inflammasome. NLRP3 was induced in brown adipocyte precursors (BAPs) from pink1 knockout (KO) mice. Unexpectedly, NLRP3 induction did not induce canonical inflammasome activity. Instead, NLRP3 induction led to the differentiation of pink1 KO BAPs into white-like adipocytes by increasing the expression of white adipocyte-specific genes and repressing the expression of brown adipocyte-specific genes. nlrp3 deletion in pink1 knockout mice reversed BAT dysfunction. Conversely, adipose tissue-specific atg7 KO mice showed significantly lower expression of Nlrp3 in their BAT. Overall, our data suggest that the role of mitophagy is different from general autophagy in regulating adipose tissue and whole-body energy metabolism. Our results uncovered a new mitochondria-NLRP3 pathway that induces BAT dysfunction. The ability of the nlrp3 knockouts to rescue BAT dysfunction suggests the transcriptional function of NLRP3 as an unexpected, but a quite specific therapeutic target for obesity-related metabolic diseases. Abbreviations: ACTB: actin, beta; BAPs: brown adipocyte precursors; BAT: brown adipose tissue; BMDMs: bone marrow-derived macrophages; CASP1: caspase 1; CEBPA: CCAAT/enhancer binding protein (C/EBP), alpha; ChIP: chromatin immunoprecipitation; EE: energy expenditure; HFD: high-fat diet; IL1B: interleukin 1 beta; ITT: insulin tolerance test; KO: knockout; LPS: lipopolysaccharide; NLRP3: NLR family, pyrin domain containing 3; PINK1: PTEN induced putative kinase 1; PRKN: parkin RBR E3 ubiquitin protein ligase; RD: regular diet; ROS: reactive oxygen species; RT: room temperature; UCP1: uncoupling protein 1 (mitochondrial, proton carrier); WT: wild-type.
- Published
- 2021
34. EPHA3 Contributes to Epigenetic Suppression of PTEN in Radioresistant Head and Neck Cancer
- Author
-
Daseul Seong, Seong-Who Kim, Hyo-Won Chang, Myung-Woul Han, Hyoung-Uk Je, Hee-Jeong Cha, Song Hee Kim, Jae-Hee An, Sang Yoon Kim, Won-Hyeok Lee, and Byung-Chul Kang
- Subjects
0301 basic medicine ,DNA (Cytosine-5-)-Methyltransferase 1 ,PTEN ,Biology ,Epigenetic Repression ,Microbiology ,Biochemistry ,Radiation Tolerance ,Article ,03 medical and health sciences ,Histone H3 ,0302 clinical medicine ,Cell Line, Tumor ,Histone methylation ,Humans ,Enhancer of Zeste Homolog 2 Protein ,EZH2 ,histone methylation ,Epigenetics ,Molecular Biology ,Protein kinase B ,DNMT1 ,Receptor, EphA3 ,PTEN Phosphohydrolase ,DNA Methylation ,QR1-502 ,radioresistance ,C-myc ,Histone Code ,030104 developmental biology ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,DNA methylation ,Cancer research ,biology.protein ,head and neck cancer ,EPHA3 - Abstract
EPHA3, a member of the EPH family, is overexpressed in various cancers. We demonstrated previously that EPHA3 is associated with radiation resistance in head and neck cancer via the PTEN/Akt/EMT pathway, the inhibition of EPHA3 significantly enhances the efficacy of radiotherapy in vitro and in vivo. In this study, we investigated the mechanisms of PTEN regulation through EPHA3-related signaling. Increased DNA methyltransferase 1 (DNMT1) and enhancer of zeste homolog 2 (EZH2) levels, along with increased histone H3 lysine 27 trimethylation (H3K27me3) levels, correlated with decreased levels of PTEN in radioresistant head and neck cancer cells. Furthermore, PTEN is regulated in two ways: DNMT1-mediated DNA methylation, and EZH2-mediated histone methylation through EPHA3/C-myc signaling. Our results suggest that EPHA3 could display a novel regulatory mechanism for the epigenetic regulation of PTEN in radioresistant head and neck cancer cells.
- Published
- 2021
35. Epigenetic regulation of p62/SQSTM1 overcomes the radioresistance of head and neck cancer cells via autophagy-dependent senescence induction
- Author
-
Kyung-Chul Choi, Myungjin Lee, Myung Woul Han, Geun-Hee Lee, Hee-Bum Kang, Eun-Ju Chang, Won Hyeok Lee, Sang Yoon Kim, Seong Who Kim, Kyung-Ja Cho, Hae Yun Nam, and Gi-Jun Sung
- Subjects
DNA (Cytosine-5-)-Methyltransferase 1 ,Male ,Cancer Research ,Methyltransferase ,Cancer therapy ,Immunology ,Mice, Nude ,Histone Deacetylase 1 ,Biology ,medicine.disease_cause ,Radiation Tolerance ,Article ,Epigenesis, Genetic ,Genetic Heterogeneity ,Cellular and Molecular Neuroscience ,Cell Line, Tumor ,Radioresistance ,Sequestosome-1 Protein ,Autophagy ,medicine ,Animals ,Humans ,Epigenetics ,lcsh:QH573-671 ,Promoter Regions, Genetic ,Cellular Senescence ,Cancer ,Squamous Cell Carcinoma of Head and Neck ,lcsh:Cytology ,Acetylation ,Cell Biology ,DNA Methylation ,Chromatin Assembly and Disassembly ,Xenograft Model Antitumor Assays ,Chromatin ,Gene Expression Regulation, Neoplastic ,Histone ,Head and Neck Neoplasms ,DNA methylation ,Cancer cell ,biology.protein ,Cancer research ,CpG Islands ,Carcinogenesis ,Signal Transduction - Abstract
Tumors are composed of subpopulations of cancer cells with functionally distinct features. Intratumoral heterogeneity limits the therapeutic effectiveness of cancer drugs. To address this issue, it is important to understand the regulatory mechanisms driving a subclonal variety within a therapy-resistant tumor. We identified tumor subclones of HN9 head and neck cancer cells showing distinct responses to radiation with different levels of p62 expression. Genetically identical grounds but epigenetic heterogeneity of the p62 promoter regions revealed that radioresistant HN9-R clones displayed low p62 expression via the creation of repressive chromatin architecture, in which cooperation between DNMT1 (DNA methyltransferases 1) and HDAC1 (histone deacetylases 1) resulted in DNA methylation and repressive H3K9me3 and H3K27me3 marks in the p62 promoter. Combined inhibition of DNMT1 and HDAC1 by genetic depletion or inhibitors enhanced the suppressive effects on proliferative capacity and in vivo tumorigenesis following irradiation. Importantly, ectopically p62-overexpressed HN9-R clones increased the induction of senescence along with p62-dependent autophagy activation. These results demonstrate the heterogeneous expression of p62 as the key component of clonal variation within a tumor against irradiation. Understanding the epigenetic diversity of p62 heterogeneity among subclones allows for improved identification of the functional state of subclones and provides a novel treatment option to resolve resistance to current therapies.
- Published
- 2021
36. Tristetraprolin Posttranscriptionally Downregulates TRAIL Death Receptors
- Author
-
Seong Who Kim, Jae Hee An, Won Hyeok Lee, Song Hee Kim, Daseul Seong, Sang Yoon Kim, Hyo Won Chang, Jong Cheol Lee, and Myung Woul Han
- Subjects
0301 basic medicine ,Untranslated region ,Tristetraprolin ,Article ,cancer treatment ,TNF-Related Apoptosis-Inducing Ligand ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,medicine ,Humans ,RNA Processing, Post-Transcriptional ,posttranscriptional modification ,Receptor ,lcsh:QH301-705.5 ,AU-rich element ,Messenger RNA ,Chemistry ,tristetraprolin ,tumor necrosis factor-related apoptosis-inducing ligand ,Cancer ,General Medicine ,AU-rich elements ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,030104 developmental biology ,lcsh:Biology (General) ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,death receptor ,Tumor necrosis factor alpha - Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has attracted attention as a potential candidate for cancer therapy. However, many primary cancers are resistant to TRAIL, even when combined with standard chemotherapy. The mechanism of TRAIL resistance in cancer cells has not been fully elucidated. The TRAIL death receptor (DR) 3&prime, untranslated region (3&prime, UTR) is reported to contain AU-rich elements (AREs) that are important for regulating DR mRNA stability. However, the mechanisms by which DR mRNA stability is determined by its 3&prime, UTR are unknown. We demonstrate that tristetraprolin (TTP), an ARE-binding protein, has a critical function of regulating DR mRNA stability. DR4 mRNA contains three AREs and DR5 mRNA contains four AREs in 3&prime, UTR. TTP bound to all three AREs in DR4 and ARE3 in DR5 and enhanced decay of DR4/5 mRNA. TTP overexpression in colon cancer cells changed the TRAIL-sensitive cancer cells to TRAIL-resistant cells, and down-regulation of TTP increased TRAIL sensitivity via DR4/5 expression. Therefore, this study provides a molecular mechanism for enhanced levels of TRAIL DRs in cancer cells and a biological basis for posttranscriptional modification of TRAIL DRs. In addition, TTP status might be a biomarker for predicting TRAIL response when a TRAIL-based treatment is used for cancer.
- Published
- 2020
37. p53-dependent glutamine usage determines susceptibility to oxidative stress in radioresistant head and neck cancer cells
- Author
-
Seong Who Kim, Mi Ra Kim, Hae Yun Nam, Jung Je Park, Song Hee Kim, Yoon Sun Lee, Sang Yoon Kim, Myungjin Lee, Myung Woul Han, Jong Cheol Lee, and Hyo Won Chang
- Subjects
0301 basic medicine ,Cell Survival ,Glutamine ,Antineoplastic Agents ,medicine.disease_cause ,Radiation Tolerance ,03 medical and health sciences ,0302 clinical medicine ,Glutaminase ,Radioresistance ,Cell Line, Tumor ,medicine ,Humans ,chemistry.chemical_classification ,Reactive oxygen species ,Chemistry ,Cancer ,Cell Biology ,medicine.disease ,Glutathione ,Oxidative Stress ,030104 developmental biology ,Glucose ,Anaerobic glycolysis ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Tumor Suppressor Protein p53 ,Reactive Oxygen Species ,Glycolysis ,Oxidation-Reduction ,Oxidative stress ,Biogenesis ,NADP ,Signal Transduction - Abstract
The manner in which p53 maintains redox homeostasis and the means by which two key metabolic elements, glucose and glutamine, contribute to p53-dependent redox stability remain unclear. To elucidate the manner in which p53 deals with glucose-deprived, reactive oxygen species (ROS)-prone conditions in this regard, two isogenic cancer subclones (HN3R-A and HN3R-B) bearing distinct p53 mutations as an in vitro model of intratumoral p53 heterogeneity were identified. Following cumulative irradiation, the subclones showed a similar metabolic shift to aerobic glycolysis and increasing NADPH biogenesis for cellular defense against oxidative damage irrespective of p53 status. The radioresistant cancer cells became more sensitive to glycolysis-targeting drugs. However, in glucose-deprived and ROS-prone conditions, HN3R-B, the subclone with the original p53 increased the utilization of glutamine by GLS2, thereby maintaining redox homeostasis and ATP. Conversely, HN3R-A, the p53-deficient radioresistant subclone displayed an impairment in glutamine usage and high susceptibility to metabolic stresses as well as ROS-inducing agents despite the increased ROS scavenging system. Collectively, our findings suggest that p53 governs the alternative utilization of metabolic ingredients, such as glucose and glutamine, in ROS-prone conditions. Thus, p53 status may be an important biomarker for selecting cancer treatment strategies, including metabolic drugs and ROS-inducing agents, for recurrent cancers after radiotherapy.
- Published
- 2020
38. EphA3 maintains radioresistance in head and neck cancers through epithelial mesenchymal transition
- Author
-
Won Hyeok Lee, Sang Yoon Kim, Song Hee Kim, Hyoung Uk Je, Hyo Won Chang, Seong Who Kim, K.-P. Kim, Myung Woul Han, Jong Cheol Lee, and Young Min Kim
- Subjects
Male ,0301 basic medicine ,Epithelial-Mesenchymal Transition ,medicine.medical_treatment ,Transplantation, Heterologous ,Mice, Nude ,Radiation Tolerance ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,Radioresistance ,medicine ,Animals ,Humans ,Epithelial–mesenchymal transition ,Radiosensitivity ,RNA, Small Interfering ,business.industry ,Receptor, EphA3 ,Head and neck cancer ,Erythropoietin-producing hepatocellular (Eph) receptor ,Receptor Protein-Tyrosine Kinases ,Cell Biology ,medicine.disease ,Radiation therapy ,030104 developmental biology ,Gamma Rays ,Head and Neck Neoplasms ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,RNA Interference ,Neoplasm Recurrence, Local ,business - Abstract
Radiotherapy is a well-established therapeutic modality used in the treatment of many cancers. However, radioresistance remains a serious obstacle to successful treatment. Radioresistance can cause local recurrence and distant metastases in some patients after radiation treatment. Thus, many studies have attempted to identify effective radiosensitizers. Eph receptor functions contribute to tumor development, modulating cell-cell adhesion, invasion, neo-angiogenesis, tumor growth and metastasis. However, the role of EphA3 in radioresistance remains unclear. In the current study, we established a stable radioresistant head and neck cancer cell line (AMC HN3R cell line) and found that EphA3 was expressed predominantly in the radioresistant head and neck cancer cell line through DNA microarray, real time PCR and Western blotting. Additionally, we found that EphA3 was overexpressed in recurrent laryngeal cancer specimens after radiation therapy. EphA3 mediated the tumor invasiveness and migration in radioresistant head and neck cancer cell lines and epithelial mesenchymal transition- related protein expression. Inhibition of EphA3 enhanced radiosensitivity in the AMC HN 3R cell line in vitro and in vivo study. In conclusion, our results suggest that EphA3 is overexpressed in radioresistant head and neck cancer and plays a crucial role in the development of radioresistance in head and neck cancers by regulating the epithelial mesenchymal transition pathway.
- Published
- 2018
39. Radiotherapy-assisted tumor selective metronomic oral chemotherapy
- Author
-
Ji Won Kim, Seung Woo Chung, Ok Cheol Jeon, Hanul Lee, Young Seok Cho, Seong Who Kim, Seho Kweon, Beom Suk Lee, In San Kim, Sang Yoon Kim, Youngro Byun, Jeong Uk Choi, Gui Chul Kim, and Foyez Mahmud
- Subjects
0301 basic medicine ,Cancer Research ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Cancer ,Prodrug ,Pharmacology ,medicine.disease ,Targeted therapy ,Radiation therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,In vivo ,Oral administration ,030220 oncology & carcinogenesis ,medicine ,Doxorubicin ,business ,medicine.drug - Abstract
Chemotherapy have commonly been used in maximum tolerated dose to completely eradicate the cancer. However, such treatments often failed due to the complex and dynamic nature of cancer. Therefore, it has been suggested that cancer should be treated as a chronic disease, controlling its growth by providing continuous therapeutic pressure for long-term. Such an approach, however, requires a therapy that is non-toxic and orally available with sufficient potency. Herein, we propose a radiotherapy-assisted orally available metronomic apoptosis-targeted chemotherapy, which delivers doxorubicin continuously to the irradiated tumor with high selectivity while causing minimal toxicities to the normal tissues. DEVD-S-DOX/DCK complex is the anticancer prodrug for our strategy that could selectively release doxorubicin in the irradiated tumor tissue with sufficient oral bioavailability. The prodrug was completely inactive by itself, but displayed potent anticancer activity when coupled with radiotherapy. Consequently, the daily oral administration of DEVD-S-DOX/DCK in combination with the low-dose radiotherapy effectively suppressed the growth of tumor in vivo with no significant systemic toxicities despite that the accumulated dose of doxorubicin exceeded 150 mg/kg. Therefore, the our novel therapy using DEVD-S-DOX/DCK complex is considered as an outstanding treatment option for treating cancer for long-term attributed to its oral availability and low-toxicity profile as well as the potent anticancer effect.
- Published
- 2017
40. Phosphorylation of p62 by AMP-activated protein kinase mediates autophagic cell death in adult hippocampal neural stem cells
- Author
-
Seong Who Kim, Kyungrim Yi, Seong-Woon Yu, Caroline Jeeyeon Hong, Eun Kyoung Kim, Seol-Hwa Jeong, Kyung Min Chung, and Shinwon Ha
- Subjects
0301 basic medicine ,Cell Survival ,Recombinant Fusion Proteins ,Nerve Tissue Proteins ,AMP-Activated Protein Kinases ,Autophagy-Related Protein 7 ,Hippocampus ,Biochemistry ,03 medical and health sciences ,Neural Stem Cells ,AMP-activated protein kinase ,Sequestosome-1 Protein ,Mitophagy ,Autophagy ,Animals ,Humans ,Point Mutation ,Phosphorylation ,Protein kinase A ,Protein Kinase Inhibitors ,Molecular Biology ,Cells, Cultured ,Tissue homeostasis ,biology ,AMPK ,Cell Biology ,Neural stem cell ,Rats ,Cell biology ,Enzyme Activation ,Adult Stem Cells ,Protein Transport ,030104 developmental biology ,Amino Acid Substitution ,Protein Synthesis and Degradation ,biology.protein ,RNA Interference ,Stem cell ,Protein Processing, Post-Translational ,Gene Deletion - Abstract
In the adult brain, programmed death of neural stem cells is considered to be critical for tissue homeostasis and cognitive function and is dysregulated in neurodegeneration. Previously, we have reported that adult rat hippocampal neural (HCN) stem cells undergo autophagic cell death (ACD) following insulin withdrawal. Because the apoptotic capability of the HCN cells was intact, our findings suggested activation of unique molecular mechanisms linking insulin withdrawal to ACD rather than apoptosis. Here, we report that phosphorylation of autophagy-associated protein p62 by AMP-activated protein kinase (AMPK) drives ACD and mitophagy in HCN cells. Pharmacological inhibition of AMPK or genetic ablation of the AMPK α2 subunit by clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 genome editing suppressed ACD, whereas AMPK activation promoted ACD in insulin-deprived HCN cells. We found that following insulin withdrawal AMPK phosphorylated p62 at a novel site, Ser-293/Ser-294 (in rat and human p62, respectively). Phosphorylated p62 translocated to mitochondria and induced mitophagy and ACD. Interestingly, p62 phosphorylation at Ser-293 was not required for staurosporine-induced apoptosis in HCN cells. To the best of our knowledge, this is the first report on the direct phosphorylation of p62 by AMPK. Our data suggest that AMPK-mediated p62 phosphorylation is an ACD-specific signaling event and provide novel mechanistic insight into the molecular mechanisms in ACD.
- Published
- 2017
41. Enhanced axonal regeneration by transplanted Wnt3a-secreting human mesenchymal stem cells in a rat model of spinal cord injury
- Author
-
Jeong Hoon Kim, Seong Who Kim, Sang Ryong Jeon, Eun-Sil Shin, Hyung Ho Yoon, Joongkee Min, and Dong Kwang Seo
- Subjects
0301 basic medicine ,animal structures ,medicine.medical_treatment ,Mesenchymal Stem Cell Transplantation ,Rats, Sprague-Dawley ,Andrology ,03 medical and health sciences ,0302 clinical medicine ,Wnt3A Protein ,medicine ,Animals ,Humans ,Spinal cord injury ,Cells, Cultured ,Spinal Cord Injuries ,Spinal Cord Regeneration ,Stem cell transplantation for articular cartilage repair ,business.industry ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Stem-cell therapy ,Anatomy ,medicine.disease ,Nerve Regeneration ,Rats ,Transplantation ,030104 developmental biology ,Spinal Cord ,embryonic structures ,Female ,Surgery ,Neurology (clinical) ,Stem cell ,business ,Microtubule-Associated Proteins ,030217 neurology & neurosurgery ,Adult stem cell - Abstract
While pure mesenchymal stem cell (MSC) treatment for spinal cord injury (SCI) is known to be safe, its efficacy is insufficient. Therefore, gene-modified stem cells are being developed to enhance the effect of pure MSCs. We investigated the effect of stem cell therapy through the transfection of a Wnt3a-producing gene that stimulates axonal regeneration. MSCs obtained from the human umbilical cord blood (hMSCs) were multiplied, cultivated, and transfected with the pLenti-Wnt3a-GFP viral vector to produce Wnt3a-secreting hMSCs. A total of 50 rats were injured with an Infinite Horizon impactor at the level of the T7-8 vertebrae. Rats were divided into five groups according to the transplanted material: (1) phosphate-buffered saline injection group (sham group, n = 10); (Pertz et al. Proc Natl Acad Sci USA 105:1931–1936, 39) Wnt3a protein injection group (Wnt3a protein group, n = 10); (3) hMSC transplantation group (MSC group, n = 10); (4) hMSCs transfected with the pLenti vector transplantation group (pLenti-MSC group, n = 10); (5) hMSCs transfected with the pLenti+Wnt3a vector transplantation group (Wnt3a-MSC group, n = 10). Behavioral tests were performed daily for the first 3 days after injury and then weekly for 8 weeks. The injured spinal cords were extracted, and axonal regeneration markers including choline acetyltransferase (ChAT), growth-associated protein 43 (GAP43), and microtubule-associated protein 2 (MAP2) were investigated by immunofluorescence, RT-PCR, and western blotting. Seven weeks after the transplantation (8 weeks after SCI), rats in the Wnt3a-MSC group achieved significantly higher average scores in the motor behavior tests than those in the other groups (p
- Published
- 2017
42. Homotypic Interaction of Stabilin-2 Plays a Critical Role in Lymph Node Metastasis of Tongue Cancer
- Author
-
In San Kim, Kyung Ja Cho, Seong Who Kim, Seung Yoon Park, Young Min Kim, Soon Yuhl Nam, Myung Woul Han, Sang Yoon Kim, Jong Cheol Lee, and Myungjin Lee
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,Cell Adhesion Molecules, Neuronal ,government.form_of_government ,Metastasis ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Tongue ,medicine ,Humans ,Tongue Neoplasm ,Lymph node ,Aged ,business.industry ,Cancer ,General Medicine ,Middle Aged ,medicine.disease ,Tongue Neoplasms ,Lymphatic Endothelium ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Lymphatic Metastasis ,030220 oncology & carcinogenesis ,Cancer research ,government ,Biomarker (medicine) ,Immunohistochemistry ,Female ,business - Abstract
Background/aim Lymph node (LN) metastasis of solid types of tumors has important clinical significance and it is therefore critical to identify molecular biomarkers that would enable the selection of patients with LN metastases. Patients and methods We evaluated the expression of stabilin-2 in primary oral tongue tumors and metastatic LNs using immunohistochemical staining. The correlation between risk factors and nodal metastasis was assessed and disease-free survival was analyzed. Results Stabilin-2 expression remained a significant predictor of LN metastasis and the factor affecting recurrence in tongue cancer. Most importantly, all metastatic tumors of tongue, lung, stomach and colon cancers stained positive for stabilin-2 and stabilin-2 was expressed strongly in the sinusoidal endothelial cell of metastatic LNs. Conclusion Stabilin-2 can play a critical role in the first entrapping step of LN metastasis through homotypic interaction with the lymphatic endothelium and appears to be a tumor biomarker predicting for LN metastasis in patients with solid tumors.
- Published
- 2016
43. The role of CIP2A as a therapeutic target of rapamycin in radioresistant head and neck cancer with TP53 mutation
- Author
-
Hae Yun Nam, Song Hee Kim, Daseul Seong, Jae Hee An, Sang Yoon Kim, Seong Who Kim, Myung Woul Han, Hyoung Uk Je, and Won Hyeok Lee
- Subjects
0301 basic medicine ,Senescence ,Male ,Cell Culture Techniques ,Autoantigens ,Radiation Tolerance ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,In vivo ,Radioresistance ,Cell Line, Tumor ,Medicine ,Animals ,Humans ,Radiosensitivity ,Cytotoxicity ,Sirolimus ,Antibiotics, Antineoplastic ,business.industry ,Squamous Cell Carcinoma of Head and Neck ,Intracellular Signaling Peptides and Proteins ,Membrane Proteins ,030104 developmental biology ,Otorhinolaryngology ,Cell culture ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Cancer cell ,Mutation ,Cancer research ,Tumor Suppressor Protein p53 ,business ,Neoplasm Transplantation - Abstract
BACKGROUND CIP2A may activate multiple oncogenic proteins and promote the proliferation of various cancer cells. METHODS We investigated that the role of CIP2A in radioresistant head and neck cancer (HNC) cell line with TP53 mutation and the effect of the rapamycin on the response of HN31 with TP53 mutation cells to irradiation related to CIP2A expression. RESULTS CIP2A expression was stimulated by p53 mutation and critical for the inhibition of senescence induction in response to radiation. The treatment with radiation alone neither induced cytotoxicity in HN31 cells nor completely suppressed the activation of CIP2A. However, the combination of radiation and rapamycin increase the radiosensitivity through the induction of senescence with downregulation of CIP2A expression both in vivo and in vitro. CONCLUSION Our results suggest that CIP2A may serve as a therapeutic target of rapamycin through induction of senescence in radioresistant HNC with TP53 mutation.
- Published
- 2019
44. MicroRNA expression profiling of adult hippocampal neural stem cells upon cell death reveals an autophagic cell death-like pattern
- Author
-
Seong Who Kim, Ha-Na Woo, Heuiran Lee, Won Il Lee, Steven Hyun Seung Lee, S. S. Park, and Rachelle Choi
- Subjects
0301 basic medicine ,Programmed cell death ,Biophysics ,Biology ,Biochemistry ,Hippocampus ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Neural Stem Cells ,microRNA ,Autophagy ,Animals ,Insulin ,Molecular Biology ,Cell Death ,Gene Expression Profiling ,Cell Biology ,Neural stem cell ,Cell biology ,Rats ,Gene expression profiling ,Adult Stem Cells ,MicroRNAs ,030104 developmental biology ,Apoptosis ,030220 oncology & carcinogenesis ,Stem cell ,Transcriptome - Abstract
Adult hippocampal neural (HCN) stem cells promptly undergo irreversible autophagic cell death (ACD) if deprived of insulin in culture. Small, non-coding microRNAs (miRNA) play an important role in regulating biological processes, including proliferation and cell death. However, there have been no reports thus far regarding miRNA involvement in the induction of adult HCN stem cell death under insulin-deprived conditions, for which we performed a microarray-based analysis to examine the expression signature of miRNAs in adult rat HCN stem cells. Three independent specimens per culture condition either with or without insulin were prepared and a miRNA microarray analysis carried out. A total of 12 exhibited significantly altered expression levels upon cell death due to the absence of insulin when compared to HCN stem cells cultured with insulin present (cut-off limit; p 0.05 and fold-change1.3) The resulting volcano plot showed that, among these miRNAs, seven were upregulated and five were downregulated. The upregulated miRNAs were capable of modulating HCN stem cell death. Caspase-3 activity analysis, LC3 conversion, and TEM of autophagosome formation consistently suggested that ACD, not apoptosis, was most likely the mechanism affecting HCN cell death. As such, we have come to term these miRNAs, "HCN stem cell-specific autophagic cell death regulators." Taken together, our data suggest that the miRNA expression profile of HCN stem cells is altered during ACD occurring due to insulin deprivation and that differentially expressed miRNAs are involved in HCN stem cell viability. Detailed explorations of the underlying mechanisms regarding HCN stem cell viability modulation by these miRNAs would be beneficial in further understanding the physiological features of adult HCN stem cells and are currently being investigated.
- Published
- 2018
45. Optimal Ratio of Wnt3a Expression in Human Mesenchymal Stem Cells Promotes Axonal Regeneration in Spinal Cord Injured Rat Model.
- Author
-
Hyung Ho Yoon, Hyang Ju Lee, Joongkee Min, Jeong Hoon Kim, Jin Hoon Park, Ji Hyun Kim, Seong Who Kim, Heuiran Lee, and Sang Ryong Jeon
- Subjects
NERVOUS system regeneration ,HUMAN stem cells ,MESENCHYMAL stem cells ,ANIMAL disease models ,SPINAL cord - Abstract
Objective: Through our previous clinical trials, the demonstrated therapeutic effects of MSC in chronic spinal cord injury (SCI) were found to be not sufficient. Therefore, the need to develop stem cell agent with enhanced efficacy is increased. We transplanted enhanced Wnt3asecreting human mesenchymal stem cells (hMSC) into injured spines at 6 weeks after SCI to improve axonal regeneration in a rat model of chronic SCI. We hypothesized that enhanced Wnt3a protein expression could augment neuro-regeneration after SCI. Methods: Thirty-six Sprague-Dawley rats were injured using an Infinite Horizon (IH) impactor at the T9-10 vertebrae and separated into five groups: 1) phosphate-buffered saline injection (injury only group, n=7); 2) hMSC transplantation (MSC, n=7); 3) hMSC transfected with pLenti vector (without Wnt3a gene) transplantation (pLenti-MSC, n=7); 4) hMSC transfected with Wnt3a gene transplantation (Wnt3a-MSC, n=7); and 5) hMSC transfected with enhanced Wnt3a gene (1.7 fold Wnt3a mRNA expression) transplantation (1.7 Wnt3a-MSC, n=8). Six weeks after SCI, each 5×10
5 cells/15 µL at 2 points were injected using stereotactic and microsyringe pump. To evaluate functional recovery from SCI, rats underwent Basso-Beattie-Bresnahan (BBB) locomotor test on the first, second, and third days post-injury and then weekly for 14 weeks. Axonal regeneration was assessed using growth-associated protein 43 (GAP43), microtubule-associated protein 2 (MAP2), and neurofilament (NF) immunostaining. Results: Fourteen weeks after injury (8 weeks after transplantation), BBB score of the 1.7 Wnt3a-MSC group (15.0±0.28) was significantly higher than that of the injury only (10.0±0.48), MSC (12.57±0.48), pLenti-MSC (12.42±0.48), and Wnt3a-MSC (13.71±0.61) groups (p<0.05). Immunostaining revealed increased expression of axonal regeneration markers GAP43, MAP2, and NF in the Wnt3a-MSC and 1.7 Wnt3a-MSC groups. Conclusion: Our results showed that enhanced gene expression of Wnt3a in hMSC can potentiate axonal regeneration and improve functional recovery in a rat model of chronic SCI. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
46. Radiotherapy-associated Furin Expression and Tumor Invasiveness in Recurrent Laryngeal Cancer
- Author
-
Sang Yoon Kim, Hyo Won Chang, Myungjin Lee, Chang Hwan Ryu, Gui Chul Kim, and Seong Who Kim
- Subjects
Male ,musculoskeletal diseases ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Epithelial-Mesenchymal Transition ,medicine.medical_treatment ,Mice, Nude ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Internal medicine ,Carcinoma ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Epithelial–mesenchymal transition ,Laryngeal Neoplasms ,Furin ,Tissue microarray ,biology ,business.industry ,Cancer ,General Medicine ,Laryngeal Neoplasm ,medicine.disease ,Matrix Metalloproteinases ,Radiation therapy ,Laryngectomy ,030104 developmental biology ,030220 oncology & carcinogenesis ,embryonic structures ,biology.protein ,Neoplasm Recurrence, Local ,business - Abstract
Background/aim Recurrent laryngeal cancer often shows an aggressive phenotype after radiotherapy and does not respond to conventional therapeutic strategies. In this study, we investigated the contribution of furin to cellular invasiveness in radio-resistant laryngeal cancer. Materials and methods Using previously established AMC-HN-3 and AMC-HN-8 cell lines from laryngeal carcinoma patients, recurrent laryngeal cancer models were generated by cumulative irradiation (AMC-HN-3-70Gy and AMC-HN-8-70Gy). Immunocytochemistry and western blotting were used to determine the epithelial-mesenchymal transition (EMT). Invasion capacity was assessed using an in vitro invasion assay. Zymography was used to assess metalloproteinase-2 (MMP-2) activity. Tumor xenografts were developed to compare growth rate and furin expression in vivo. Furin expression in 35 patients (45 samples) with salvage total laryngectomy after radiation-based treatment was assessed by laryngeal cancer tissue microarray. Results Both AMC-HN-3-70Gy and AMC-HN-8-70Gy cell lines underwent EMT following radiation. However, AMC-HN-3-70Gy cells showed increased cellular invasiveness, whereas AMC-HN-8-70Gy cells showed no difference. AMC-HN-3-70Gy cells also exhibited elevated furin expression with up-regulated expression of the active form of membrane type 1-matrix metalloproteinase (MT1-MMP)/MMP-2, whereas AMC-HN-8-70Gy cells did not show significant changes. After administration of a furin inhibitor (chloromethyl ketone (CMK)), AMC-HN-3-70Gy cells showed a significant decrease in MT1-MMP/MMP-2 expression and cellular invasiveness. Nine of 22 samples (40.9%) from salvage total laryngectomy and one of 13 pre-radiation samples (7.7%) had high furin expression. Post-radiation, furin expression increased in seven of 10 patients whose pre- and post-radiation samples were available; all-cancer mortality (three patients) was observed in this group. Conclusion Together with EMT, furin activity may serve as an indicator of an aggressive cancer phenotype, suggesting that furin is a potentially useful target for recurrent laryngeal cancer.
- Published
- 2016
47. Albumin-binding caspase-cleavable prodrug that is selectively activated in radiation exposed local tumor
- Author
-
Seung Woo Chung, Seong Who Kim, Ok Cheol Jeon, Youngro Byun, Sang Yoon Kim, Julia Byun, In San Kim, Jeong Uk Choi, and Beom Seok Lee
- Subjects
Male ,medicine.medical_treatment ,Biophysics ,Bioengineering ,Caspase 3 ,02 engineering and technology ,Pharmacology ,Targeted therapy ,Biomaterials ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Albumins ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Prodrugs ,Doxorubicin ,Caproates ,Caspase ,Cell Proliferation ,biology ,Cell growth ,business.industry ,Prodrug ,021001 nanoscience & nanotechnology ,Up-Regulation ,Mechanics of Materials ,Apoptosis ,030220 oncology & carcinogenesis ,Drug delivery ,Ceramics and Composites ,biology.protein ,Peptides ,0210 nano-technology ,business ,medicine.drug - Abstract
Existence of the genomically and epigenomically diverse subclones in a tumor severely limits the therapeutic efficacy of targeted agents. To overcome such a limitation, we prepared a novel targeted prodrug, EMC-DEVD-S-DOX, which comprises two important features: radiation-induced apoptosis targeting and albumin-binding properties. In particular, the prodrug binds circulating albumin after intravenous administration and then activated by caspase-3, which is upregulated from apoptotic cells that responded to radiotherapy. The prodrug was designed to bind circulating albumin to extend half-life and facilitate tumor accumulation in order to increase the possibility of contacting caspase-3, which is only transiently upregulated during apoptosis. Our results showed that EMC-DEVD-S-DOX had a prolonged half-life with enhanced tumor accumulation, which clearly benefited the therapeutic effect of the prodrug. Also, agreeing with the in vitro studies that showed ignorable cytotoxic effect in the absence of caspase-3, the prodrug was effective only when combined with radiotherapy without any noticeable systemic toxicity in vivo. Due to the highly selective action of EMC-DEVD-S-DOX independent to the complex genomic profiles of tumor, the prodrug would overcome the limitation of current targeted therapy and potentiate radiotherapy in the clinical oncology.
- Published
- 2016
48. The Effects of Losartan in Preserving the Structural Integrity of Decellularized Small Diameter Vascular Allograft Conduit Implants In Vivo
- Author
-
Seong Who Kim, Seung Hyun Lee, Suk Jung Choo, and Byoung Wook Lee
- Subjects
0301 basic medicine ,Intimal hyperplasia ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,030204 cardiovascular system & hematology ,Biomaterials ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Blood vessel prosthesis ,In vivo ,Medicine ,DAPI ,Decellularization ,business.industry ,General Medicine ,medicine.disease ,030104 developmental biology ,Losartan ,medicine.anatomical_structure ,chemistry ,Immunohistochemistry ,business ,Elastic fiber ,Biomedical engineering ,medicine.drug - Abstract
Decellularization is a proposed method of preparing nonautologous biological arterial vascular scaffolding; however, the fate of the supporting medial elastic fiber, which is important in preserving the vascular structural integrity, is uncertain. The influence of losartan on preserving the medial elastic fiber integrity in decellularized small diameter vascular conduits (SDVC) was investigated. Decellularized infrarenal abdominal aortic allografts were implanted in Sprague-Dawley rats treated either with (study rats, n = 6) or without oral losartan (control rats, n = 6) and graded 8 weeks later according to a remodeling scoring system (1-mild, 2-moderate, 3-severe) which we devised based on the intimal hyperplasia degree, morphologic changes, and elastic fiber fragmentation of the conduits. DAPI immunohistochemistry analysis was performed in 47 (25 decellularization only and 22 losartan treatment) cross-sectional slide specimens. The losartan versus decellularization only SDVC showed a significantly lower medial elastic fragmentation score (1.32 vs. 2.24, P < 0.001), superior medial layer preservation, and relatively more normal appearing intimal cellular morphology. The results suggested rats receiving decellularized SDVCs treated with losartan may yield superior medial layer elastic fiber preservation.
- Published
- 2016
49. Senescence-Associated MCP-1 Secretion Is Dependent on a Decline in BMI1 in Human Mesenchymal Stromal Cells
- Author
-
Min Kyung Kim, Seong Who Kim, Soo Jin Choi, Jisun Lim, Dong-Myung Shin, Gyong Hwa Hong, You Sook Cho, Hyang Ju Lee, Mi-Yeon Kim, Hae Yun Nam, Jinbeom Heo, In Gyu Kim, and Hye Jin Jin
- Subjects
0301 basic medicine ,Senescence ,CCR2 ,Transcription, Genetic ,Receptors, CCR2 ,Physiology ,Clinical Biochemistry ,Protein Array Analysis ,Paracrine Communication ,Biology ,p38 Mitogen-Activated Protein Kinases ,Biochemistry ,03 medical and health sciences ,Paracrine signalling ,Animals ,Humans ,Secretion ,Autocrine signalling ,Molecular Biology ,Cells, Cultured ,Cellular Senescence ,Chemokine CCL2 ,General Environmental Science ,Polycomb Repressive Complex 1 ,fungi ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cell Biology ,Fetal Blood ,Asthma ,Cell biology ,Autocrine Communication ,Disease Models, Animal ,Oxidative Stress ,Original Research Communications ,Phenotype ,030104 developmental biology ,Cancer research ,Cytokines ,General Earth and Planetary Sciences ,Cell aging ,Protein Binding - Abstract
Aims: Cellular senescence and its secretory phenotype (senescence-associated secretory phenotype [SASP]) develop after long-term expansion of mesenchymal stromal cells (MSCs). Further investigation of this phenotype is required to improve the therapeutic efficacy of MSC-based cell therapies. In this study, we show that positive feedback between SASP and inherent senescence processes plays a crucial role in the senescence of umbilical cord blood-derived MSCs (UCB-MSCs). Results: We found that monocyte chemoattractant protein-1 (MCP-1) was secreted as a dominant component of the SASP during expansion of UCB-MSCs and reinforced senescence via its cognate receptor chemokine (c-c motif) receptor 2 (CCR2) by activating the ROS-p38-MAPK-p53/p21 signaling cascade in both an autocrine and paracrine manner. The activated p53 in turn increased MCP-1 secretion, completing a feed-forward loop that triggered the senescence program in UCB-MSCs. Accordingly, knockdown of CCR2 in UCB-MSCs significantly improved their therapeutic ability to alleviate airway inflammation in an experimental allergic asthma model. Moreover, BMI1, a polycomb protein, repressed the expression of MCP-1 by binding to its regulatory elements. The reduction in BMI1 levels during UCB-MSC senescence altered the epigenetic status of MCP-1, including the loss of H2AK119Ub, and resulted in derepression of MCP-1. Innovation: Our results provide the first evidence supporting the existence of the SASP as a causative contributor to UCB-MSC senescence and reveal a so far unappreciated link between epigenetic regulation and SASP for maintaining a stable senescent phenotype. Conclusion: Senescence of UCB-MSCs is orchestrated by MCP-1, which is secreted as a major component of the SASP and is epigenetically regulated by BMI1. Antioxid. Redox Signal. 24, 471–485.
- Published
- 2016
50. Targeting prion-like protein doppel selectively suppresses tumor angiogenesis
- Author
-
Seung Woo Chung, Youngro Byun, Taslim A. Al-Hilal, Jeong Uk Choi, Sang Yoon Kim, Jooho Park, Seong Who Kim, Farzana Alam, In San Kim, and Fakhrul Ahsan
- Subjects
Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Prions ,Angiogenesis ,media_common.quotation_subject ,medicine.medical_treatment ,Mice, Nude ,Antineoplastic Agents ,Biology ,GPI-Linked Proteins ,Cell membrane ,Neovascularization ,Mice ,03 medical and health sciences ,Drug Delivery Systems ,0302 clinical medicine ,Neoplasms ,medicine ,Animals ,Humans ,Internalization ,media_common ,Mice, Inbred BALB C ,Neovascularization, Pathologic ,Growth factor ,Endothelial Cells ,Kinase insert domain receptor ,Hep G2 Cells ,General Medicine ,Vascular Endothelial Growth Factor Receptor-2 ,Xenograft Model Antitumor Assays ,In vitro ,Neoplasm Proteins ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,cardiovascular system ,Female ,Signal transduction ,medicine.symptom ,Research Article ,Signal Transduction - Abstract
Controlled and site-specific regulation of growth factor signaling remains a major challenge for current antiangiogenic therapies, as these antiangiogenic agents target normal vasculature as well tumor vasculature. In this article, we identified the prion-like protein doppel as a potential therapeutic target for tumor angiogenesis. We investigated the interactions between doppel and VEGFR2 and evaluated whether blocking the doppel/VEGFR2 axis suppresses the process of angiogenesis. We discovered that tumor endothelial cells (TECs), but not normal ECs, express doppel; tumors from patients and mouse xenografts expressed doppel in their vasculatures. Induced doppel overexpression in ECs enhanced vascularization, whereas doppel constitutively colocalized and complexed with VEGFR2 in TECs. Doppel inhibition depleted VEGFR2 from the cell membrane, subsequently inducing the internalization and degradation of VEGFR2 and thereby attenuating VEGFR2 signaling. We also synthesized an orally active glycosaminoglycan (LHbisD4) that specifically binds with doppel. We determined that LHbisD4 concentrates over the tumor site and that genetic loss of doppel in TECs decreases LHbisD4 binding and targeting both in vitro and in vivo. Moreover, LHbisD4 eliminated VEGFR2 from the cell membrane, prevented VEGF binding in TECs, and suppressed tumor growth. Together, our results demonstrate that blocking doppel can control VEGF signaling in TECs and selectively inhibit tumor angiogenesis.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.