11 results on '"Sae Bom Lee"'
Search Results
2. A Novel IL2Rα-/- Model for CAR-T Toxicity in Acute Lymphoblastic Leukemia Recapitulates Cytokine Release Syndrome and Neutropenia
- Author
-
Payal Goala, Hiroshi Kotani, Zhang Yongliang, Kayla M. Reid, Justin C. Boucher, Constanza Savid-Frontera, Sae Bom Lee, Nolan Beatty, Michael D. Jain, and Marco L. Davila
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
3. The Regulation of NRF2 by Nutrient-Responsive Signaling and Its Role in Anabolic Cancer Metabolism
- Author
-
Sae Bom Lee, Brianna N Sellers, and Gina M DeNicola
- Subjects
0301 basic medicine ,NF-E2-Related Factor 2 ,Physiology ,Somatic cell ,medicine.medical_treatment ,Clinical Biochemistry ,Biology ,medicine.disease_cause ,digestive system ,environment and public health ,Biochemistry ,Antioxidants ,03 medical and health sciences ,Neoplasms ,medicine ,Animals ,Humans ,Molecular Biology ,Transcription factor ,Cell Proliferation ,General Environmental Science ,Kelch-Like ECH-Associated Protein 1 ,Growth factor ,Nutrients ,Cell Biology ,respiratory system ,KEAP1 ,Cell biology ,030104 developmental biology ,Cancer cell ,General Earth and Planetary Sciences ,Signal transduction ,Carcinogenesis ,Oxidative stress ,Signal Transduction - Abstract
The stress responsive transcription factor nuclear factor erythroid 2 p45-related factor 2, or NRF2, regulates the expression of many cytoprotective enzymes to mitigate oxidative stress under physiological conditions. NRF2 is activated in response to oxidative stress, growth factor signaling, and changes in nutrient status. In addition, somatic mutations that disrupt the interaction between NRF2 and its negative regulator Kelch-like erythroid cell-derived protein with CNC homology (ECH)-associated 1 (KEAP1) commonly occur in cancer and are thought to promote tumorigenesis. Recent Advances: While it is well established that aberrant NRF2 activation results in enhanced antioxidant capacity in cancer cells, recent exciting findings demonstrate a role for NRF2-mediated metabolic deregulation that supports cancer cell proliferation.In this review, we describe how the NRF2-KEAP1 signaling pathway is altered in cancer, how NRF2 is regulated by changes in cellular metabolism, and how NRF2 reprograms cellular metabolism to support proliferation.Future studies will delineate the NRF2-regulated processes critical for metabolic adaptation to nutrient availability, cellular proliferation, and tumorigenesis. Antioxid. Redox Signal. 00, 000-000.
- Published
- 2018
- Full Text
- View/download PDF
4. Metabolically engineered glucose-utilizing Shewanella strains under anaerobic conditions
- Author
-
In Geol Choi, Sohyun Kim, In Seop Chang, Byoungnam Min, Donggeon Choi, and Sae Bom Lee
- Subjects
Shewanella ,Environmental Engineering ,Bioelectric Energy Sources ,Bioengineering ,Zymomonas mobilis ,Microbiology ,Electricity ,Glucokinase ,Glycolysis ,Anaerobiosis ,Shewanella oneidensis ,Waste Management and Disposal ,chemistry.chemical_classification ,biology ,Strain (chemistry) ,Renewable Energy, Sustainability and the Environment ,General Medicine ,PEP group translocation ,biology.organism_classification ,Aerobiosis ,Glucose ,Enzyme ,Metabolic Engineering ,chemistry ,Biochemistry - Abstract
Comparative genome analysis of Shewanella strains predicted that the strains metabolize preferably two- and three-carbon carbohydrates as carbon/electron source because many Shewanella genomes are deficient of the key enzymes in glycolysis (e.g., glucokinase). In addition, all Shewanella genomes are known to have only one set of genes associated with the phosphotransferase system required to uptake sugars. To engineer Shewanella strains that can utilize five- and six-carbon carbohydrates, we constructed glucose-utilizing Shewanella oneidensis MR-1 by introducing the glucose facilitator (glf; ZMO0366) and glucokinase (glk; ZMO0369) genes of Zymomonas mobilis. The engineered MR-1 strain was able to grow on glucose as a sole carbon/electron source under anaerobic conditions. The glucose affinity (Ks) and glucokinase activity in the engineered MR-1 strain were 299.46 mM and 0.259 ± 0.034 U/g proteins. The engineered strain was successfully applied to a microbial fuel cell system and exhibited current generation using glucose as the electron source.
- Published
- 2014
- Full Text
- View/download PDF
5. MDSC Suppression of CAR T Cells Can be Reduced By Targeted Signaling Disruption
- Author
-
Sae Bom Lee, Justin C. Boucher, Kayla Reid, Estelle V Cervantes, Kristen Spitler, and Marco L. Davila
- Subjects
Tumor microenvironment ,LAG3 ,medicine.medical_treatment ,T cell ,Immunology ,CD28 ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Immune system ,Cytokine ,medicine.anatomical_structure ,Antigen ,Myeloid-derived Suppressor Cell ,Cancer research ,medicine - Abstract
CAR T cells are genetically modified with an extracellular scFv, transmembrane domain, and intracellular costimulatory and CD3ζ domains. Two treatments received the approval from the FDA for the treatment of acute lymphoblastic leukemia and diffuse large B cell lymphoma. However, CAR T cell persistence remains a problem. A reason for this may be that myeloid cells such as myeloid derived suppressor cells (MDSCs) may be contributing to the reduced persistence of CAR T cells. MDSCs originate from myeloid cells and have been implicated in the suppression of the immune system in the tumor microenvironment. To determine what effect MDSCs might have during CAR T cell production we co-cultured MDSCs during CAR transduction. We found gene transfer was lower for m19z (38.7% vs 46.8%), m1928z (24.1% vs 39.1%), and m19hBBz (35.8% vs 46.2%) CAR T cells co-cultured with MDSCs compared to those that were not (Fig 1A). There was also a reduction in total T cell counts for m19z (58%), m1928z (88%), and m19hBBz (65%) after MDSC co-culture. This data suggests MDSCs present during CAR T cell production can alter gene transfer and total T cell counts. We also investigated the effect MDSCs can potentially have on CAR T cells when present during CAR T cell antigen stimulation. CAR T cells co-cultured with MDSCs in vitro had significant reductions in m19z, m1928z, and m19hBBz CAR T cell mediated killing against target cells (Fig 1B). We also found significantly lower production of IFNγ in m19z, m1928z, and m19hBBz CAR T cells cultured with MDSCs compared to cells cultured with total BM (Fig 1C). After 24hr stimulation with MDSCs and target cells, there was lower expression of activation markers PD1 and LAG3 by CAR T cells compared to culture without MDSCs. This suggests that MDSCs reduce CAR T cell activation, killing, and cytokine production. We also looked at how CAR proliferation after antigen stimulation is affected by MDSC co-culture and found MDSCs significantly reduced CAR proliferation in vitro. To evaluate if we could create a more resistant CAR T cell to MDSC suppression null mutations were incorporated into a CD28 CAR. We mutated the YMNM and PRRP subdomains of CD28 which signal through PI3K and ITK respectively leaving only PYAP active (mut06). When MDSCs were co-cultured with mut06 T cells during production mut06 had a smaller reduction in gene transfer (21% vs 38%) and T cell counts (80% vs 88%) compared to m1928z. Mut06 also had a significantly higher expression of PD1 and TIM3 compared to m1928z after production with MDSCs. In vitro when mut06 was co-cultured with MDSCs it had the same killing ability as m1928z without MDSCs and was significantly better at killing compared to m1928z co-cultured with MDSCs (Fig 1B). To examine the effect of MDSCs on CAR T cells in vivo we injected C57BL6 mice with CAR T cells followed by MDSCs a week later. In these ongoing experiments we found mut06 had significantly higher numbers of CAR T cells in the blood compared to m1928z (Fig1D). Overall our data shows that MDSCs can suppress CAR T cell function when present during production as well as CAR stimulation. It also suggests that by optimizing CD28 CAR signaling using mut06 that we were able to generate a CAR T cell that is more resistant to MDSCs. Furthermore, we may be able to recapitulate the effect of mut06 with targeted inhibitors. Figure 1 Disclosures Davila: GlaxoSmithKline: Consultancy; Novartis: Research Funding; Adaptive: Consultancy; Celgene: Research Funding; Precision Biosciences: Consultancy; Bellicum: Consultancy; Anixa: Consultancy; Atara: Research Funding.
- Published
- 2019
- Full Text
- View/download PDF
6. Tumor Inflammation and Myeloid Derived Suppressor Cells Reduce the Efficacy of CD19 CAR T Cell Therapy in Lymphoma
- Author
-
Mohammad Hussaini, Hiroshi Kotani, Justin C. Boucher, Frederick L. Locke, John E. Mullinax, Meghan Menges, Crystal R Pope, Christina A Bachmeier, Ricardo J. Gonzalez, Julio C. Chavez, Hua Zhao, Michael D. Jain, Reginald Atkins, Rawan Faramand, Bijal D. Shah, Sae Bom Lee, and Marco L. Davila
- Subjects
Oncology ,medicine.medical_specialty ,education.field_of_study ,Myeloid ,business.industry ,Immunology ,Population ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Lymphoma ,medicine.anatomical_structure ,Specimen collection ,Internal medicine ,medicine ,Myeloid-derived Suppressor Cell ,Bone marrow ,Sample collection ,education ,B-cell lymphoma ,business - Abstract
Introduction: Approximately 60% of Large B cell Lymphoma (LBCL) patients that receive CD19 CAR T cell therapy with axicabtagene ciloleucel (axi-cel) experience lymphoma progression (Locke et al. Lancet Oncol. 2019) and the likelihood of response to subsequent therapy is low (Spiegel, Dahiya et al. ASCO 2019). Target loss of CD19 is observed in less than a third of patients experiencing relapse. Alternative mechanisms of resistance to axi-cel are poorly understood. Lymphoma patients with elevated serum markers of systemic inflammation, such as ferritin and IL-6, have worse outcomes following axi-cel (Locke, Neelapu et al. Mol.Ther.2017; Faramand et al. ASH 2018). We hypothesized that suppressive monocytic myeloid derived suppressor cells (M-MDSCs), which are associated with worse chemotherapy outcomes in LBCL (Azzaoui et al. Blood 2016), and tumor driven inflammation may be present and responsible for decreased efficacy of axi-cel in LBCL. Methods: LBCL patients undergoing axi-cel treatment were enrolled onto prospective sample collection protocols. Patients were stratified for analysis into ongoing responders (complete response or partial response) or relapsed (progressive disease) after a minimum of 3 months follow-up (range 3 - 15 months). M-MDSCs, defined as a Lin-, CD11b+, CD33+, CD15-, CD14+, HLA-DRlow population, were sorted from leftover apheresis material after collection for axi-cel manufacture. M-MDSC ability to suppress proliferation of autologous T cells stimulated with CD3/CD28 coated beads was measured by 3H thymidine incorporation. Circulating peripheral blood M-MDSCs, quantified by % of live cells by flow cytometry, were measured at the time of apheresis and serially after axi-cel infusion until day 30. In vitro mouse experiments utilized a CD19-CD28 CAR and cytokine-induced bone marrow MDSCs (Thevenot et al. Immunity 2014). Cytokines were measured by ELISA and cytotoxicity against CD19 bearing cell lines used xCELLigence real-time cell analysis, as we have done previously (Li et al. JCI Insight 2018).Tumor biopsies were taken within 1 month prior to infusion of axi-cel. Limited gene expression profiling of tumor microenvironment (TME) genes used the Nanostring IO360 panel (770 genes). Analysis used nSolver to identify cell types, GSEA and differential gene expression between groups. Results: First, we demonstrated that M-MDSCs sorted from patient apheresis material suppressed the proliferation of autologous T cells (n=6). We next enumerated M-MDSCs in the peripheral blood (n = 32). M-MDSC numbers initially decreased after lymphodepleting chemotherapy but recovered to baseline levels by day +10. The level of M-MDSCs following CAR T cell therapy strongly correlated with pre-CAR T baseline levels (R = 0.871, p Conclusions: Systemic inflammatory myeloid cytokines, circulating M-MDSCs in the blood and chronic IFN in the TME all associate with LBCL relapse after axi-cel CAR T cell therapy. Our observations support that CAR T cells can be suppressed by baseline patient and tumor-related factors and strategies to overcome these factors should be targeted to improve patient outcomes. MDJ and HZ contributed equally. Disclosures Jain: Kite/Gilead: Consultancy. Bachmeier:Kite/Gilead: Speakers Bureau. Chavez:Novartis: Membership on an entity's Board of Directors or advisory committees; Genentech: Speakers Bureau; Kite Pharmaceuticals, Inc.: Membership on an entity's Board of Directors or advisory committees; Janssen Pharmaceuticals, Inc.: Speakers Bureau. Shah:Jazz Pharmaceuticals: Research Funding; Incyte: Research Funding; Kite/Gilead: Honoraria; Celgene/Juno: Honoraria; Pharmacyclics: Honoraria; Adaptive Biotechnologies: Honoraria; Spectrum/Astrotech: Honoraria; Novartis: Honoraria; AstraZeneca: Honoraria. Mullinax:Iovance: Research Funding. Davila:Celgene: Research Funding; GlaxoSmithKline: Consultancy; Precision Biosciences: Consultancy; Novartis: Research Funding; Atara: Research Funding; Bellicum: Consultancy; Adaptive: Consultancy; Anixa: Consultancy. Locke:Kite: Other: Scientific Advisor; Novartis: Other: Scientific Advisor; Cellular BioMedicine Group Inc.: Consultancy.
- Published
- 2019
- Full Text
- View/download PDF
7. Mutation of the CD28 Co-Stimulatory Domain Confers Enhanced CAR T Cell Function
- Author
-
Maria L. Cabral, Marco L. Davila, Said M. Sebti, Dylan Morrissey, Justin C. Boucher, Kristen Spitler, Xuefeng Wang, Aslamuzzaman Kazi, Sae Bom Lee, Gongbo Li, Bishwas Shrestha, Hiroshi Kotani, Bin Yu, and Nolan J. Beatty
- Subjects
LAG3 ,T cell ,Immunology ,CD28 ,NFAT ,Cell Biology ,Hematology ,Biology ,HAVCR2 ,Biochemistry ,CD19 ,Cell biology ,medicine.anatomical_structure ,medicine ,biology.protein ,Signal transduction ,human activities ,Transcription factor - Abstract
An obstacle with continued clinical development of CAR T cells is the limited understanding of their biology and mechanisms of anti-tumor immunity. We and others have shown that CARs with a CD28 co-stimulatory domain drive high levels of T cell activation that also lead to exhaustion and shortened persistence. The CD28 domain includes 3 intracellular subdomains (YMNM, PRRP, and PYAP) that regulate signaling pathways post TCR-stimulation, but it is unknown how they modulate activation and/or exhaustion of CAR T cells. A detailed understanding of the mechanism of CD28-dependent exhaustion in CAR T cells will allow the design of a CAR less prone to exhaustion and reduce relapse rates. This led us to hypothesize that by incorporating null mutations of CD28 subdomains (Fig 1A) we could optimize CAR T cell signaling and reduce exhaustion. In vitro, we found mutated CAR T cells with only a functional PYAP (mut06) subdomain secrete significantly less IFNγ, IL6, and TNFα after 24hr stimulation compared to non-mutated CD28 CAR T cells, but greater than the 1st generation m19z CAR. Also, cytotoxicity was enhanced compared to non-mutated CARs (Fig 1B). Using a pre-clinical immunocompetent mouse tumor model, we found the mut06 CAR T cell treated mice had a significant survival advantage compared to non-mutated CD28 CAR T cells (Fig 1C). To examine exhaustion, we ex vivo stimulated CAR T cells with target cells expressing CD19 and PDL1 and found mut06 CAR T cells had increased IFNγ (42%), TNFα (62%) and IL2 (73%) secretion compared to exhausted non-mutated CD28 CAR T cells. This suggests that mut06 CAR T cells are more resistant to exhaustion. To find a mechanistic explanation for this observation we examined CAR T cell signaling. After 24hr stimulation with CD19 target cells mut06 CAR T cells had a significant reduction in pAkt compared to m1928z CAR T cells, which is a critical signaling mediator in the NFAT and NR4A1 transcription factor pathways. Additionally, mut06 had decreased p-NFAT compared to m1928z when examined by western blot. To determine how optimized CAR signaling affected T cell exhaustion we looked at 22 genes that are upregulated when NFAT is constitutively active and overlap with genes identified as important for T cell exhaustion. We found that most of the exhaustion related genes were upregulated in m1928z CAR T cells while they were decreased in m19hBBz. The mut06 CAR T cell gene expression pattern was more similar to m19hBBz with exhaustion related genes downregulated compared to m1928z (Fig 1D). To examine differences in the accessibility of exhaustion related genes we performed ATAC-seq and found NFAT (Nfatc1) and NR4A2 (Nr4a2) had lower chromatin accessibility profiles in mut06 compared to m1928z (Fig 1E). We also found that exhaustion related genes Havcr2 (TIM3), Pdcd1 (PD1), and Lag3 (LAG3) all had greatly reduced chromatin accessibility in mut06 CAR T cells compared m1928z. Overall, these genomic studies support our findings that mut06 optimizes CAR T cell signaling by lowering transcription factors that regulate exhaustion. Figure 1 Disclosures Li: ImmuneBro Therapeutics: Other: sole shareholder . Davila:Atara: Research Funding; Celgene: Research Funding; GlaxoSmithKline: Consultancy; Novartis: Research Funding; Anixa: Consultancy; Bellicum: Consultancy; Adaptive: Consultancy; Precision Biosciences: Consultancy.
- Published
- 2019
- Full Text
- View/download PDF
8. The Lymphoma Tumor Microenvironment Influences Toxicity after CD19 CAR T Cell Therapy
- Author
-
Bijal D. Shah, Julio C. Chavez, Marco L. Davila, John E. Mullinax, Michael D. Jain, Verena Staedtke, Brigett Brandjes, Rawan Faramand, Kayla Reid, Hiroshi Kotani, Mohammad Hussaini, Frederick L. Locke, Christina A Bachmeier, Renyuan Bai, Sae Bom Lee, Kristen Spitler, Ricardo J. Gonzalez, Xuefeng Wang, and Marian Dam
- Subjects
0301 basic medicine ,Adoptive cell transfer ,business.industry ,medicine.medical_treatment ,T cell ,Immunology ,FOXP3 ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,03 medical and health sciences ,Cytokine release syndrome ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Cytokine ,Interleukin 15 ,medicine ,Cytokine secretion ,business ,B cell ,030215 immunology - Abstract
Introduction: Of patients receiving CD19 CAR T cell therapy for large B cell lymphoma (LBCL), approximately 1 in 10 experience severe cytokine release syndrome (CRS) and 1 in 3 experience severe neurotoxicity. While CAR T cells trigger the onset of these toxicities, CRS and neurotoxicity are thought to occur as a consequence of activated myeloid cells amplifying cytokine and catecholamine release, thereby stimulating inflammation both systemically and at the blood-brain barrier. However, patient and tumor-related factors that account for differences in the amount of toxicity remain poorly understood. Methods: Serum cytokine levels were measured on an ELLA point of care device prior to lymphodepleting chemotherapy and throughout inpatient treatment with CD19 CAR T cell therapy (axicabtagene ciloleucel) for LBCL. Catecholamine levels were measured as we have previously reported. Tumor biopsies were taken within 1 month prior to infusion of CAR T cells. RNA expression was measured by RNAseq and/or a Nanostring IO360 panel consisting of 770 genes found in the tumor microenvironment (TME) in cancer. Analysis used nSolver to identify cell types, GSEA and differential gene expression between groups. Mouse CAR T cell studies utilized mouse CD19-targeted CAR T cells derived from C57BL/6 splenocytes and cultured in vitro with myeloid cells and target cells to evaluate cytotoxicity and/or cytokine secretion. Elicited mouse macrophages were collected from peritoneal fluid 4 days after IP injection of 3% Brewer's thioglycollate medium. In vivo studies with mouse CD19-targeted CAR T cells were performed in IL2Ra-/- mice given cyclophosphamide as a pre-conditioning chemotherapy followed by adoptive transfer and analyses for CAR T cell and B cell persistence, as well as cytokines. Results: Of 58 patients undergoing CD19 CAR T cell therapy for LBCL, 8 (14%) had severe (grade 3 or higher) CRS and 16 (28%) had severe (grade 3 or higher) neurotoxicity. At baseline, peripheral blood levels of IL-6, IFN-γ, IL-15 and ferritin were significantly higher in patients who would subsequently experience severe CRS and severe neurotoxicity. Confirming our recent animal model of CRS we determined that peak serum catecholamine levels were higher in patients experiencing severe CRS. To identify if myeloid cells potentiate cytokine release we co-cultured CAR T cells with CD19 target and macrophages obtained from elicited mouse peritoneum. When these macrophages were added, IL-6 release from CAR T cells significantly increased compared to when macrophages were absent. Next, we studied the baseline TME in LBCL CAR T patients. Of 36 patients, 10 (27%) experienced severe neurotoxicity following CAR T cell therapy. By cell type score, the severe neurotoxicity group had a lower expression of genes associated with T cells overall and specifically Tregs. Also significantly lower in the severe neurotoxicity group were T cell genes including multiple subunits of CD3, CD3ζ, FOXP3, ICOS, CD62L and others. Association of increased T cell infiltration in the TME with low neurotoxicity raised the possibility that suppressive T cell subsets play a role in limiting toxicity post-CAR T cell therapy. To test this hypothesis, we injected CD19-targeted CAR T cells into an immune competent mouse model of Treg depletion (IL2Ra-/-) with established CD19+ leukemia. Treg deficient mice experienced a massive cytokine release after CAR T infusion and died prematurely due to CAR T toxicity compared to control mice with Tregs intact. Conclusions: Our observations suggest that the incidence of severe toxicity following CD19 CAR T cell therapy is influenced by baseline characteristics that are present prior to the infusion of CAR T cells. These include systemic inflammation characterized by high cytokine levels and a TME notable for a lack of infiltrating T cells. We posit a model whereby inflammation primes myeloid cells that are further activated upon CAR T cell infusion to release toxic amounts of cytokines and catecholamines. T cell subsets in the TME may modulate CAR T cells at the site of antigen encounter and prevent excessive CAR T activation. Reducing systemic inflammation or encouraging T cell infiltration into tumor prior to CAR T infusion are potential strategies for lowering the toxicity associated with CAR T therapy. Disclosures Jain: Kite/Gilead: Consultancy. Chavez:Kite Pharmaceuticals, Inc.: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Genentech: Speakers Bureau; Janssen Pharmaceuticals, Inc.: Speakers Bureau. Shah:Novartis: Honoraria; Spectrum/Astrotech: Honoraria; Celgene/Juno: Honoraria; Kite/Gilead: Honoraria; Incyte: Research Funding; Jazz Pharmaceuticals: Research Funding; Pharmacyclics: Honoraria; Adaptive Biotechnologies: Honoraria; AstraZeneca: Honoraria. Bachmeier:Kite/Gilead: Speakers Bureau. Mullinax:Iovance: Research Funding. Locke:Novartis: Other: Scientific Advisor; Cellular BioMedicine Group Inc.: Consultancy; Kite: Other: Scientific Advisor. Davila:Anixa: Consultancy; Precision Biosciences: Consultancy; Novartis: Research Funding; GlaxoSmithKline: Consultancy; Adaptive: Consultancy; Celgene: Research Funding; Atara: Research Funding; Bellicum: Consultancy.
- Published
- 2019
- Full Text
- View/download PDF
9. Metabolic Profiling Regarding Pathogenesis of Idiopathic Pulmonary Fibrosis
- Author
-
Do Jin Kim, Hwa Kyun Shin, Chang Woo Choi, Choon-Sik Park, Yun Pyo Kang, Eun Suk Koh, Ji Min Lee, Ji Yeon Hong, Sung Won Kwon, Hyung Min Kim, Sae Bom Lee, Sung Woo Park, and Won Jun Lee
- Subjects
0301 basic medicine ,Metabolite ,Ornithine aminotransferase ,Biology ,Biochemistry ,Gas Chromatography-Mass Spectrometry ,Pathogenesis ,03 medical and health sciences ,chemistry.chemical_compound ,Idiopathic pulmonary fibrosis ,0302 clinical medicine ,Metabolomics ,Fibrosis ,medicine ,Humans ,Myofibroblasts ,Cells, Cultured ,Lung ,Discriminant Analysis ,General Chemistry ,respiratory system ,medicine.disease ,Idiopathic Pulmonary Fibrosis ,respiratory tract diseases ,Metabolic pathway ,030104 developmental biology ,medicine.anatomical_structure ,030228 respiratory system ,chemistry ,Case-Control Studies ,Immunology ,Cancer research ,Metabolic Networks and Pathways - Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, eventually fatal disease characterized by fibrosis of the lung parenchyma and loss of lung function. IPF is believed to be caused by repetitive alveolar epithelial cell injury and dysregulated repair process including uncontrolled proliferation of lung (myo) fibroblasts and excessive deposition of extracellular matrix proteins in the interstitial space; however, the pathogenic pathways involved in IPF have not been fully elucidated. In this study, we attempted to characterize metabolic changes of lung tissues involved in the pathogenesis of IPF using gas chromatography-mass spectrometry-based metabolic profiling. Partial least-squares discriminant analysis (PLS-DA) model generated from metabolite data was able to discriminate between the control subjects and IPF patients (R(2)X = 0.37, R(2)Y = 0.613 and Q(2) (cumulative) = 0.54, receiver operator characteristic AUC0.9). We discovered 25 metabolite signatures of IPF using both univariate and multivariate statistical analyses (FDR0.05 and VIP score of PLS-DA1). These metabolite signatures indicated alteration in metabolic pathways: adenosine triphosphate degradation pathway, glycolysis pathway, glutathione biosynthesis pathway, and ornithine aminotransferase pathway. The results could provide additional insight into understanding the disease and potential for developing biomarkers.
- Published
- 2016
10. Novel approach for analysis of bronchoalveolar lavage fluid (BALF) using HPLC-QTOF-MS-based lipidomics: lipid levels in asthmatics and corticosteroid-treated asthmatic patients
- Author
-
Sung Won Kwon, Jeong Hill Park, Donghak Kim, Sae Bom Lee, Choon-Sik Park, Won Jun Lee, Sunghyouk Park, Yun Pyo Kang, Sung Woo Park, and Ji Yeon Hong
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,medicine.drug_class ,Anti-Inflammatory Agents ,Biochemistry ,Mass Spectrometry ,chemistry.chemical_compound ,Young Adult ,Adrenal Cortex Hormones ,Internal medicine ,Lipidomics ,medicine ,Humans ,Respiratory system ,Chromatography, High Pressure Liquid ,Phospholipids ,Triglycerides ,Asthma ,Triglyceride ,medicine.diagnostic_test ,Computational Biology ,General Chemistry ,Phosphatidylserine ,Middle Aged ,medicine.disease ,respiratory tract diseases ,Endocrinology ,Bronchoalveolar lavage ,chemistry ,Immunology ,Corticosteroid ,lipids (amino acids, peptides, and proteins) ,Female ,Sphingomyelin ,Bronchoalveolar Lavage Fluid - Abstract
To better understand the respiratory lipid phenotypes of asthma, we developed a novel method for lipid profiling of bronchoalveolar lavage fluid (BALF) using HPLC-QTOF-MS with an internal spectral library and high-throughput lipid-identifying software. The method was applied to BALF from 38 asthmatic patients (18 patients with nonsteroid treated bronchial asthma [NSBA] and 20 patients with steroid treated bronchial asthma [SBA]) and 13 healthy subjects (NC). We identified 69 lipids, which were categorized into one of six lipid classes: lysophosphatidylcholine (LPC), phosphatidylcholine (PC), phosphatidylglycerol (PG), phosphatidylserine (PS), sphingomyelin (SM) and triglyceride (TG). Compared with the NC group, the individual quantity levels of the six classes of lipids were significantly higher in the NSBA subjects. In the SBA subjects, the PC, PG, PS, SM, and TG levels were similar to the levels observed in the NC group. Using differentially expressed lipid species (p value0.05, FDR0.1 and VIP score of PLS-DA1), 34 lipid biomarker candidates with high prediction performance between asthmatics and controls were identified (AUROC0.9). These novel findings revealed specific characteristics of lipid phenotypes in asthmatic patients and suggested the importance of future research on the relationship between lipid levels and asthma.
- Published
- 2014
11. Dietary hematein ameliorates fatty streak lesions in the rabbit by the possible mechanism of reducing VCAM-1 and MCP-1 expression
- Author
-
Chul-Ho Lee, Byung Hwa Hyun, Sae Bom Lee, Tae Sook Jeong, Goo Taeg Oh, Dae Young Kim, Jae-Hoon Choi, Sei-Ryang Oh, Song Hae Bok, Ju Ryoung Kim, and Jung Joo Hong
- Subjects
Male ,Transcriptional Activation ,medicine.medical_specialty ,Endothelium ,Arteriosclerosis ,Probucol ,Vascular Cell Adhesion Molecule-1 ,Aorta, Thoracic ,Electrophoretic Mobility Shift Assay ,Biology ,Polymerase Chain Reaction ,Monocytes ,Cell Line ,chemistry.chemical_compound ,Internal medicine ,medicine ,Cell Adhesion ,Animals ,VCAM-1 ,Hematein ,Hematoxylin ,Cells, Cultured ,Chemokine CCL2 ,Caesalpinia ,Plant Extracts ,Tumor Necrosis Factor-alpha ,Monocyte ,Anticholesteremic Agents ,Fatty streak ,NF-kappa B ,Blotting, Northern ,Lipids ,Endothelial stem cell ,Lipoproteins, LDL ,Endocrinology ,medicine.anatomical_structure ,Biochemistry ,chemistry ,Human umbilical vein endothelial cell ,Endothelium, Vascular ,Rabbits ,Cardiology and Cardiovascular Medicine ,Oxidation-Reduction ,medicine.drug ,Drugs, Chinese Herbal - Abstract
Hematein is a compound isolated from Caesalpinia sappan that has been used in oriental medicine as both an analgesic and an anti-inflammatory agent. In this study, we examined the anti-atherogenic potential of hematein using cholesterol-fed New Zealand White (NZW) rabbits. NZW rabbits were divided into a hematein-supplemented (0.05% in diet) group (n=6), a probucol-supplemented (0.25% in diet) group (n=6), and a control group (n=6). After 8 weeks of treatments, the extent of the atherosclerotic lesions was significantly reduced in the hematein-supplemented group and the probucol-supplemented group without changing plasma lipoprotein levels. Hematein and probucol prevented the up-regulation of the vascular cell adhesion molecule-1 (VCAM-1) expression on the descending aorta induced by cholesterol diet. In culture, hematein also significantly inhibited the secretion of soluble VCAM-1 and of monocyte chemotactic protein-1 (MCP-1) respectively induced by tumor necrotic factor alpha (TNF-alpha) and mildly oxidized low density lipoprotein in human umbilical vein endothelial cell (HUVEC) culture. Also, hematein inhibited monocyte adhesion to endothelial cell and the activation of NF-kappaB in HUVECs stimulated with TNF-alpha. The results of the present study suggest that the anti-atherogenic effect of hematein is not related to control of the plasma lipid profile but probably related to the inhibition of VCAM-1 and MCP-1 expression resulting in an amelioration of lesion development in the rabbit.
- Published
- 2001
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.