1
|
Kim D, Yoon MS, Lee J, Park SY, Han JS. Effects of phospholipase D1-inhibitory peptide on the growth and metastasis of gastric cancer cells. Mol Cells 2024; 47:100128. [PMID: 39426685 PMCID: PMC11582423 DOI: 10.1016/j.mocell.2024.100128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024] Open
Abstract
Phospholipase D1 (PLD1) contributes to cancer development and progression through its effects on cell proliferation, survival, invasion, metastasis, angiogenesis, drug resistance, and modulation of the tumor microenvironment. Its central role in these processes makes it a promising target for novel cancer treatments aimed at inhibiting its activity and disrupting the signaling pathways it regulates. In this study, we aimed to investigate the effect of PLD1 inhibition on gastric cancer cell growth using a novel peptide inhibitor, TAT-TVTSP. PLD1, which plays a role in cancer progression, catalyzes the conversion of phosphatidylcholine into choline and phosphatidic acid through hydrolysis. To effectively target PLD1 in cells, we engineered TAT-TVTSP by fusing a PLD1-inhibitory peptide (TVTSP) with a cell-penetrating peptide (TAT). We observed that TAT-TVTSP effectively inhibited PLD1 activity in AGS gastric cancer cells. Moreover, TAT-TVTSP significantly inhibited the mammalian target of the rapamycin signaling pathway, including the phosphorylation of key downstream targets such as S6K1, AKT, S473, glycogen synthase kinase-3b, and forkhead box O1. TAT-TVTSP did not induce cell death, but it triggered cell cycle arrest by activating p21 and p27 via AKT phosphorylation. Functional assays revealed that TAT-TVTSP significantly impaired the colony-forming ability of AGS cells, thus inhibiting cell proliferation. Transwell and wound-healing assays revealed that this peptide disrupted the cellular behaviors critical to cancer progression, such as migration and invasion. In vivo, TAT-TVTSP significantly reduced tumor growth in the xenograft model of gastric cancer without any toxicity. Overall, our results suggest that TAT-TVTSP is a novel therapeutic agent for PLD1-mediated cancers.
Collapse
Affiliation(s)
- Dongju Kim
- Department of Biomedical Sciences, Graduate School for Biomedical Science & Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Mee-Sup Yoon
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Junwon Lee
- Department of Biotechnology, Pai Chai University, Daejeon 35345, Republic of Korea
| | - Shin-Young Park
- Department of Biotechnology, Pai Chai University, Daejeon 35345, Republic of Korea.
| | - Joong-Soo Han
- Department of Biomedical Sciences, Graduate School for Biomedical Science & Engineering, Hanyang University, Seoul 04763, Republic of Korea; Biomedical Research Institute and Department of Biochemistry & Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Priyadarshani P, Van Grouw A, Liversage AR, Rui K, Nikitina A, Tehrani KF, Aggarwal B, Stice SL, Sinha S, Kemp ML, Fernández FM, Mortensen LJ. Investigation of MSC potency metrics via integration of imaging modalities with lipidomic characterization. Cell Rep 2024; 43:114579. [PMID: 39153198 DOI: 10.1016/j.celrep.2024.114579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 06/17/2024] [Accepted: 07/18/2024] [Indexed: 08/19/2024] Open
Abstract
Mesenchymal stem/stromal cell (MSC) therapies have had limited success so far in clinical trials due in part to heterogeneity in immune-responsive phenotypes. Therefore, techniques to characterize these properties of MSCs are needed during biomanufacturing. Imaging cell shape, or morphology, has been found to be associated with MSC immune responsivity-but a direct relationship between single-cell morphology and function has not been established. We used label-free differential phase contrast imaging and matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) to evaluate single-cell morphology and explore relationships with lipid metabolic immune response. In interferon gamma (IFN-γ)-stimulated MSCs, we found higher lipid abundances from the ceramide-1-phosphate (C1P), phosphatidylcholine (PC), LysoPC, and triglyceride (TAG) families that are involved in cell immune function. Furthermore, we identified differences in lipid signatures in morphologically defined MSC subpopulations. The use of single-cell optical imaging coupled with single-cell spatial lipidomics could assist in optimizing the MSC production process and improve mechanistic understanding of manufacturing process effects on MSC immune activity and heterogeneity.
Collapse
Affiliation(s)
- Priyanka Priyadarshani
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA 30602, USA; Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Alexandria Van Grouw
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Adrian Ross Liversage
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA 30602, USA; Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Kejie Rui
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA 30602, USA; Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Arina Nikitina
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Kayvan Forouhesh Tehrani
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Champaign, IL 61820, USA
| | - Bhavay Aggarwal
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
| | - Steven L Stice
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA
| | - Saurabh Sinha
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
| | - Melissa L Kemp
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
| | - Facundo M Fernández
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Luke J Mortensen
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA 30602, USA; Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
3
|
Wang Y, Wakelam MJO, Bankaitis VA, McDermott MI. The wide world of non-mammalian phospholipase D enzymes. Adv Biol Regul 2024; 91:101000. [PMID: 38081756 DOI: 10.1016/j.jbior.2023.101000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 02/25/2024]
Abstract
Phospholipase D (PLD) hydrolyses phosphatidylcholine (PtdCho) to produce free choline and the critically important lipid signaling molecule phosphatidic acid (PtdOH). Since the initial discovery of PLD activities in plants and bacteria, PLDs have been identified in a diverse range of organisms spanning the taxa. While widespread interest in these proteins grew following the discovery of mammalian isoforms, research into the PLDs of non-mammalian organisms has revealed a fascinating array of functions ranging from roles in microbial pathogenesis, to the stress responses of plants and the developmental patterning of flies. Furthermore, studies in non-mammalian model systems have aided our understanding of the entire PLD superfamily, with translational relevance to human biology and health. Increasingly, the promise for utilization of non-mammalian PLDs in biotechnology is also being recognized, with widespread potential applications ranging from roles in lipid synthesis, to their exploitation for agricultural and pharmaceutical applications.
Collapse
Affiliation(s)
- Y Wang
- Department of Cell Biology & Genetics, Texas A&M Health Science Center, College Station, TX, 77843, USA; Department of Microbiology, University of Washington, Seattle, WA98109, USA
| | - M J O Wakelam
- Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, United Kingdom
| | - V A Bankaitis
- Department of Cell Biology & Genetics, Texas A&M Health Science Center, College Station, TX, 77843, USA; Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843, USA; Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - M I McDermott
- Department of Cell Biology & Genetics, Texas A&M Health Science Center, College Station, TX, 77843, USA.
| |
Collapse
|
4
|
Hou C, Ye Z, Yang S, Jiang Z, Wang J, Wang E. Lysine demethylase 1B (Kdm1b) enhances somatic reprogramming through inducing pluripotent gene expression and promoting cell proliferation. Exp Cell Res 2022; 420:113339. [PMID: 36075448 DOI: 10.1016/j.yexcr.2022.113339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 08/28/2022] [Accepted: 08/31/2022] [Indexed: 11/28/2022]
Abstract
Lysine demethylase 1B (Kdm1b) is known as an epigenetic modifier with demethylase activity against H3K4 and H3K9 histones and plays an important role in tumor progression and tumor stem cell enrichment. In this study, we attempted to elucidate the role of Kdm1b in somatic cell reprogramming. We found that exogenous expression of Kdm1b in human dermal fibroblasts (HDFs) can influence the epigenetic modifications of histones. Subsequent analysis further suggests that the overexpression of Kdm1b can promote cell proliferation, reprogram metabolism and inhibit cell apoptosis. In addition, a series of multipotent factors including Sox2 and Nanog, and several epigenetic factors that may reduce epigenetic barriers were upregulated to varying degrees. More importantly, HDFs transfected with the combination of Oct4 (POU5F1), Sox2, Klf4 and c-Myc and Kdm1b (OSKMK) achieved higher reprogramming efficiency. Therefore, we suggest that Kdm1b is an important epigenetic factor associated with pluripotency.
Collapse
Affiliation(s)
- Cuicui Hou
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, PR China; College of Chemistry, Jilin University, Changchun, Jilin, 130021, PR China
| | - Zhikai Ye
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, PR China
| | - Songqin Yang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, PR China
| | - Zhenlong Jiang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, PR China.
| | - Jin Wang
- Department of Chemistry, Physics and Applied Mathematics, State University of New York at Stony Brook, Stony Brook, NY, 11794-3400, United States.
| | - Erkang Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, PR China; College of Chemistry, Jilin University, Changchun, Jilin, 130021, PR China.
| |
Collapse
|
5
|
Lipid metabolic Reprogramming: Role in Melanoma Progression and Therapeutic Perspectives. Cancers (Basel) 2020; 12:cancers12113147. [PMID: 33121001 PMCID: PMC7692067 DOI: 10.3390/cancers12113147] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Melanoma is a devastating skin cancer characterized by an impressive metabolic plasticity. Melanoma cells are able to adapt to the tumor microenvironment by using a variety of fuels that contribute to tumor growth and progression. In this review, the authors summarize the contribution of the lipid metabolic network in melanoma plasticity and aggressiveness, with a particular attention to specific lipid classes such as glycerophospholipids, sphingolipids, sterols and eicosanoids. They also highlight the role of adipose tissue in tumor progression as well as the potential antitumor role of drugs targeting critical steps of lipid metabolic pathways in the context of melanoma. Abstract Metabolic reprogramming contributes to the pathogenesis and heterogeneity of melanoma. It is driven both by oncogenic events and the constraints imposed by a nutrient- and oxygen-scarce microenvironment. Among the most prominent metabolic reprogramming features is an increased rate of lipid synthesis. Lipids serve as a source of energy and form the structural foundation of all membranes, but have also emerged as mediators that not only impact classical oncogenic signaling pathways, but also contribute to melanoma progression. Various alterations in fatty acid metabolism have been reported and can contribute to melanoma cell aggressiveness. Elevated expression of the key lipogenic fatty acid synthase is associated with tumor cell invasion and poor prognosis. Fatty acid uptake from the surrounding microenvironment, fatty acid β-oxidation and storage also appear to play an essential role in tumor cell migration. The aim of this review is (i) to focus on the major alterations affecting lipid storage organelles and lipid metabolism. A particular attention has been paid to glycerophospholipids, sphingolipids, sterols and eicosanoids, (ii) to discuss how these metabolic dysregulations contribute to the phenotype plasticity of melanoma cells and/or melanoma aggressiveness, and (iii) to highlight therapeutic approaches targeting lipid metabolism that could be applicable for melanoma treatment.
Collapse
|
6
|
McDermott MI, Wang Y, Wakelam MJO, Bankaitis VA. Mammalian phospholipase D: Function, and therapeutics. Prog Lipid Res 2019; 78:101018. [PMID: 31830503 DOI: 10.1016/j.plipres.2019.101018] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/08/2019] [Accepted: 10/14/2019] [Indexed: 01/23/2023]
Abstract
Despite being discovered over 60 years ago, the precise role of phospholipase D (PLD) is still being elucidated. PLD enzymes catalyze the hydrolysis of the phosphodiester bond of glycerophospholipids producing phosphatidic acid and the free headgroup. PLD family members are found in organisms ranging from viruses, and bacteria to plants, and mammals. They display a range of substrate specificities, are regulated by a diverse range of molecules, and have been implicated in a broad range of cellular processes including receptor signaling, cytoskeletal regulation and membrane trafficking. Recent technological advances including: the development of PLD knockout mice, isoform-specific antibodies, and specific inhibitors are finally permitting a thorough analysis of the in vivo role of mammalian PLDs. These studies are facilitating increased recognition of PLD's role in disease states including cancers and Alzheimer's disease, offering potential as a target for therapeutic intervention.
Collapse
Affiliation(s)
- M I McDermott
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114, United States of America.
| | - Y Wang
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114, United States of America; Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843-2128, United States of America
| | - M J O Wakelam
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - V A Bankaitis
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114, United States of America; Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843-2128, United States of America; Department of Chemistry, Texas A&M University, College Station, Texas 77840, United States of America
| |
Collapse
|
7
|
Jiang Y, Liu F, Zou F, Zhang Y, Wang B, Zhang Y, Lian A, Han X, Liu Z, Liu X, Jin M, Wang D, Li G, Liu J. PBX homeobox 1 enhances hair follicle mesenchymal stem cell proliferation and reprogramming through activation of the AKT/glycogen synthase kinase signaling pathway and suppression of apoptosis. Stem Cell Res Ther 2019; 10:268. [PMID: 31443676 PMCID: PMC6708256 DOI: 10.1186/s13287-019-1382-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND PBX homeobox 1 (PBX1) is involved in the maintenance of the pluripotency of human embryonic and hematopoietic stem cells; however, the effects of PBX1 in the self-renewal and reprogramming of hair follicle mesenchymal stem cells (HF-MSCs) are unclear. The AKT/glycogen synthase kinase (GSK) 3β pathway regulates cell metabolism, proliferation, apoptosis, and reprogramming, and p16 and p21, which act downstream of this pathway, regulate cell proliferation, cell cycle, and apoptosis induced by reprogramming. Here, we aimed to elucidate the roles of PBX1 in regulating the proliferation and reprogramming of HF-MSCs. METHODS A lentiviral vector designed to carry the PBX1 sequence or PBX1 short hairpin RNA sequence was used to overexpress or knock down PBX1. The roles of PBX1 in proliferation and apoptosis were investigated by flow cytometry. Real-time polymerase chain reaction was performed to evaluate pluripotent gene expression. Dual-luciferase reporter assays were performed to examine the transcriptional activity of the NANOG promoter. Western blotting was performed to identify the molecules downstream of PBX1 involved in proliferation and reprogramming. Caspase3 activity was detected to assess HF-MSC reprogramming. The phosphatidylinositol 3-kinase/AKT inhibitor LY294002 was used to inhibit the phosphorylation and activity of AKT. RESULTS Overexpression of PBX1 in HF-MSCs increased the phosphorylation of AKT and nuclear translocation of β-catenin, resulting in the progression of the cell cycle from G0/G1 to S phase. Moreover, transfection with a combination of five transcription factors (SOMKP) in HF-MSCs enhanced the formation of alkaline phosphatase-stained colonies compared with that in HF-MSCs transfected with a combination of four transcription factors (SOMK). PBX1 upregulated Nanog transcription by activating the promoter and promoted the expression of endogenous SOX2 and OCT4. Furthermore, PBX1 expression activated the AKT/glycogen synthase kinase (GSK) 3β pathway and reduced apoptosis during the early stages of reprogramming. Inhibition of phospho-AKT or knockdown of PBX1 promoted mitochondrion-mediated apoptosis and reduced reprogramming efficiency. CONCLUSIONS PBX1 enhanced HF-MSC proliferation, and HF-MSCs induced pluripotent stem cells (iPSC) generation by activating the AKT/GSK3β signaling pathway. During the reprogramming of HF-MSCs into HF-iPSCs, PBX1 activated the NANOG promoter, upregulated NANOG, and inhibited mitochondrion-mediated apoptosis via the AKT/GSK3β pathway during the early stages of reprogramming.
Collapse
Affiliation(s)
- Yixu Jiang
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021 China
| | - Feilin Liu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130021 China
| | - Fei Zou
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, 130021 China
| | - Yingyao Zhang
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021 China
| | - Bo Wang
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021 China
| | - Yuying Zhang
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021 China
| | - Aobo Lian
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021 China
| | - Xing Han
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021 China
| | - Zinan Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021 China
| | - Xiaomei Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021 China
| | - Minghua Jin
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021 China
| | - Dianliang Wang
- Stem Cell and Tissue Engineering Research Laboratory, PLA Rocket Force Characteristic Medical Center, Beijing, 100088 China
| | - Gang Li
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, 999077 China
| | - Jinyu Liu
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021 China
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021 China
| |
Collapse
|
8
|
Kang H, Kim H, Lee S, Youn H, Youn B. Role of Metabolic Reprogramming in Epithelial⁻Mesenchymal Transition (EMT). Int J Mol Sci 2019; 20:ijms20082042. [PMID: 31027222 PMCID: PMC6514888 DOI: 10.3390/ijms20082042] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/08/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023] Open
Abstract
Activation of epithelial–mesenchymal transition (EMT) is thought to be an essential step for cancer metastasis. Tumor cells undergo EMT in response to a diverse range of extra- and intracellular stimulants. Recently, it was reported that metabolic shifts control EMT progression and induce tumor aggressiveness. In this review, we summarize the involvement of altered glucose, lipid, and amino acid metabolic enzyme expression and the underlying molecular mechanisms in EMT induction in tumor cells. Moreover, we propose that metabolic regulation through gene-specific or pharmacological inhibition may suppress EMT and this treatment strategy may be applied to prevent tumor progression and improve anti-tumor therapeutic efficacy. This review presents evidence for the importance of metabolic changes in tumor progression and emphasizes the need for further studies to better understand tumor metabolism.
Collapse
Affiliation(s)
- Hyunkoo Kang
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - Hyunwoo Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Korea.
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
- Department of Biological Sciences, Pusan National University, Busan 46241, Korea.
| |
Collapse
|
9
|
Al-Moujahed A, Tian B, Efstathiou NE, Konstantinou EK, Hoang M, Lin H, Miller JW, Vavvas DG. Receptor interacting protein kinase 3 (RIP3) regulates iPSCs generation through modulating cell cycle progression genes. Stem Cell Res 2019; 35:101387. [PMID: 30703581 PMCID: PMC7375132 DOI: 10.1016/j.scr.2019.101387] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 12/24/2018] [Accepted: 01/18/2019] [Indexed: 01/02/2023] Open
Abstract
The molecular mechanisms involved in induced pluripotent stem cells (iPSCs) generation are poorly understood. The cell death machinery of apoptosis-inducing caspases have been shown to facilitate the process of iPSCs reprogramming. However, the effect of other cell death processes, such as programmed necrosis (necroptosis), on iPSCs induction has not been studied. In this study, we investigated the role of receptor-interacting protein kinase 3 (RIP3), an essential regulator of necroptosis, in reprogramming mouse embryonic fibroblast cells (MEFs) into iPSCs. RIP3 was found to be upregulated in iPSCs compared to MEFs. Deletion of RIP3 dramatically suppressed the reprogramming of iPSCs (~82%). RNA-seq analysis and qRT-PCR showed that RIP3 KO MEFs expressed lower levels of genes that control cell cycle progression and cell division and higher levels of extracellular matrix-regulating genes. The growth rate of RIP3 KO MEFs was significantly slower than WT MEFs. These findings can partially explain the inhibitory effects of RIP3 deletion on iPSCs generation and show for the first time that the necroptosis kinase RIP3 plays an important role in iPSC reprogramming. In contrast to RIP3, the kinase and scaffolding functions of RIPK1 appeared to have distinct effects on reprogramming.
Collapse
Affiliation(s)
- Ahmad Al-Moujahed
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, United States; Department of Pathology, Boston University School of Medicine, Boston, MA 02118, United States
| | - Bo Tian
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, United States; Department of Ophthalmology & Visual Sciences, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Nikolaos E Efstathiou
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, United States
| | - Eleni K Konstantinou
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, United States
| | - Mien Hoang
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, United States
| | - Haijiang Lin
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, United States; Department of Ophthalmology & Visual Sciences, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Joan W Miller
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, United States
| | - Demetrios G Vavvas
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, United States.
| |
Collapse
|
10
|
Xiao M, Zhong H, Xia L, Tao Y, Yin H. Pathophysiology of mitochondrial lipid oxidation: Role of 4-hydroxynonenal (4-HNE) and other bioactive lipids in mitochondria. Free Radic Biol Med 2017; 111:316-327. [PMID: 28456642 DOI: 10.1016/j.freeradbiomed.2017.04.363] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 04/21/2017] [Accepted: 04/24/2017] [Indexed: 02/06/2023]
Abstract
Mitochondrial lipids are essential for maintaining the integrity of mitochondrial membranes and the proper functions of mitochondria. As the "powerhouse" of a cell, mitochondria are also the major cellular source of reactive oxygen species (ROS). Oxidative stress occurs when the antioxidant system is overwhelmed by overproduction of ROS. Polyunsaturated fatty acids in mitochondrial membranes are primary targets for ROS attack, which may lead to lipid peroxidation (LPO) and generation of reactive lipids, such as 4-hydroxynonenal. When mitochondrial lipids are oxidized, the integrity and function of mitochondria may be compromised and this may eventually lead to mitochondrial dysfunction, which has been associated with many human diseases including cancer, cardiovascular diseases, diabetes, and neurodegenerative diseases. How mitochondrial lipids are oxidized and the underlying molecular mechanisms and pathophysiological consequences associated with mitochondrial LPO remain poorly defined. Oxidation of the mitochondria-specific phospholipid cardiolipin and generation of bioactive lipids through mitochondrial LPO has been increasingly recognized as an important event orchestrating apoptosis, metabolic reprogramming of energy production, mitophagy, and immune responses. In this review, we focus on the current understanding of how mitochondrial LPO and generation of bioactive lipid mediators in mitochondria are involved in the modulation of mitochondrial functions in the context of relevant human diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Mengqing Xiao
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS), Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Huiqin Zhong
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China; University of the Chinese Academy of Sciences, CAS, Beijing, China
| | - Lin Xia
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS), Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
| | - Yongzhen Tao
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS), Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
| | - Huiyong Yin
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS), Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; University of the Chinese Academy of Sciences, CAS, Beijing, China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China.
| |
Collapse
|
11
|
Wei R, Zhao X, Hao H, Du W, Zhu H. Embryonic stem-like cells from rabbit blastocysts cultured with melatonin could differentiate into three germ layers in vitro and in vivo. Mol Reprod Dev 2016; 83:1003-1014. [DOI: 10.1002/mrd.22739] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 09/14/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Ruxue Wei
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences (IAS); Chinese Academy of Agricultural Sciences (CAAS); Beijing P.R. China
| | - Xueming Zhao
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences (IAS); Chinese Academy of Agricultural Sciences (CAAS); Beijing P.R. China
| | - Haisheng Hao
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences (IAS); Chinese Academy of Agricultural Sciences (CAAS); Beijing P.R. China
| | - Weihua Du
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences (IAS); Chinese Academy of Agricultural Sciences (CAAS); Beijing P.R. China
| | - Huabin Zhu
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences (IAS); Chinese Academy of Agricultural Sciences (CAAS); Beijing P.R. China
| |
Collapse
|