1
|
Zhao L, Luo T, Zhang H, Fan X, Zhang Q, Chen H. Kaempferol enhances intestinal repair and inhibits the hyperproliferation of aging intestinal stem cells in Drosophila. Front Cell Dev Biol 2024; 12:1491740. [PMID: 39450272 PMCID: PMC11499188 DOI: 10.3389/fcell.2024.1491740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Intestinal stem cells (ISCs) are crucial for tissue repair and homeostasis because of their ability to self-renew and differentiate. However, their functionality declines significantly with age, resulting in reduced tissue regeneration and a higher risk of age-related diseases. Addressing this decline in ISC performance during aging presents a substantial challenge. The specific impact of nutrients or dietary elements on ISC adaptive resizing is urgent to explore. Methods Drosophila ISCs are an ideal model for studying development and aging because of their genetic richness, ease of manipulation, and similarity to mammalian tissues. As the primary mitotically active cells in the Drosophila gut, ISCs are flexible in response to dietary and stress signals. Manipulating signaling pathways or dietary restrictions has shown promise in regulating ISC functions and extending lifespan in flies, these approaches face broader applications for aging research. Results Kaempferol is well-regarded for its antioxidant, anti-inflammatory, and potential anticancer effects. However, its impacts on ISCs and the associated mechanisms remain inadequately understood. Our findings indicate that Kaempferol accelerates gut recovery after damage and improves the organism's stress tolerance. Moreover, Kaempferol suppresses the hyperproliferation of aging ISCs in Drosophila. Further investigation revealed that the regulatory effects of Kaempferol on ISCs are mediated through the reduction of endoplasmic reticulum (ER) stress in aging flies and the modulation of excessive reactive oxygen species (ROS) levels via ER-stress pathways. Furthermore, Kaempferol exerts regulatory effects on the insulin signaling pathway, thereby contributing to the attenuation of ISC senescence. Discussion This study reveals that Kaempferol promotes intestinal homeostasis and longevity in aging flies by targeting ER stress and insulin signaling pathways, though the exact molecular mechanisms require further exploration. Future research will aim to dissect the downstream signaling events involved in these pathways to better understand how Kaempferol exerts its protective effects at the molecular level.
Collapse
Affiliation(s)
- Liusha Zhao
- Center of Gerontology and Geriatrics and Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Luo
- Center of Gerontology and Geriatrics and Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Zhang
- Department of Gastroenterology and Hepatology and Laboratory of Inflammatory Bowel, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xinxin Fan
- Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiaoqiao Zhang
- Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Haiyang Chen
- Center of Gerontology and Geriatrics and Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Yan L, Guo X, Zhou J, Zhu Y, Zhang Z, Chen H. Quercetin Prevents Intestinal Stem Cell Aging via Scavenging ROS and Inhibiting Insulin Signaling in Drosophila. Antioxidants (Basel) 2022; 12:antiox12010059. [PMID: 36670921 PMCID: PMC9854609 DOI: 10.3390/antiox12010059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/11/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
Adult stem cells, a class of cells that possess self-renewal and differentiation capabilities, modulate tissue regeneration, repair, and homeostasis maintenance. These cells undergo functional degeneration during aging, resulting in decreased tissue regeneration ability and increased disease incidence. Thus, it is essential to provide effective therapeutic solutions to preventing the aging-related functional decline of stem cells. Quercetin (Que) is a popular natural polyphenolic flavonoid found in various plant species. It exhibits many beneficial effects against aging and aging-related diseases; however, its efficacy against adult stem cell aging remains largely unclear. Drosophila possesses a mammalian-like intestinal system with a well-studied intestinal stem cell (ISC) lineage, making it an attractive model for adult stem cell research. Here, we show that Que supplementation could effectively prevent the hyperproliferation of ISCs, maintain intestinal homeostasis, and prolong the lifespan in aged Drosophila. In addition, we found that Que could accelerate recovery of the damaged gut and improve the tolerance of Drosophila to stressful stimuli. Furthermore, results demonstrated that Que prevents the age-associated functional decline of ISCs via scavenging reactive oxygen species (ROS) and inhibiting the insulin signaling pathway. Overall, our findings suggest that Que plays a significant role in delaying adult stem cell aging.
Collapse
Affiliation(s)
- La Yan
- Department of Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
- Laboratory of Metabolism and Aging Research, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Xiaoxin Guo
- Laboratory of Metabolism and Aging Research, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Juanyu Zhou
- Laboratory of Metabolism and Aging Research, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yuedan Zhu
- Laboratory of Metabolism and Aging Research, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Zehong Zhang
- Laboratory of Metabolism and Aging Research, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Haiyang Chen
- Laboratory of Metabolism and Aging Research, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
- Correspondence:
| |
Collapse
|
3
|
Sim EZ, Enomoto T, Shiraki N, Furuta N, Kashio S, Kambe T, Tsuyama T, Arakawa A, Ozawa H, Yokoyama M, Miura M, Kume S. Methionine metabolism regulates pluripotent stem cell pluripotency and differentiation through zinc mobilization. Cell Rep 2022; 40:111120. [PMID: 35858556 DOI: 10.1016/j.celrep.2022.111120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 04/19/2022] [Accepted: 06/28/2022] [Indexed: 11/03/2022] Open
Abstract
Pluripotent stem cells (PSCs) exhibit a unique feature that requires S-adenosylmethionine (SAM) for the maintenance of their pluripotency. Methionine deprivation in the medium causes a reduction in intracellular SAM, thus rendering PSCs in a state potentiated for differentiation. In this study, we find that methionine deprivation triggers a reduction in intracellular protein-bound Zn content and upregulation of Zn exporter SLC30A1 in PSCs. Culturing PSCs in Zn-deprived medium results in decreased intracellular protein-bound Zn content, reduced cell growth, and potentiated differentiation, which partially mimics methionine deprivation. PSCs cultured under Zn deprivation exhibit an altered methionine metabolism-related metabolite profile. We conclude that methionine deprivation potentiates differentiation partly by lowering cellular Zn content. We establish a protocol to generate functional pancreatic β cells by applying methionine and Zn deprivation. Our results reveal a link between Zn signaling and methionine metabolism in the regulation of cell fate in PSCs.
Collapse
Affiliation(s)
- Erinn Zixuan Sim
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Takayuki Enomoto
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Nobuaki Shiraki
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan.
| | - Nao Furuta
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Soshiro Kashio
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Taiho Kambe
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Tomonori Tsuyama
- Division of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Akihiro Arakawa
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto, Kawasaki-shi, Kanagawa, Japan
| | - Hiroki Ozawa
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Mizuho Yokoyama
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto, Kawasaki-shi, Kanagawa, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shoen Kume
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan.
| |
Collapse
|
4
|
Au HK, Peng SW, Guo CL, Lin CC, Wang YL, Kuo YC, Law TY, Ho HN, Ling TY, Huang YH. Niche Laminin and IGF-1 Additively Coordinate the Maintenance of Oct-4 Through CD49f/IGF-1R-Hif-2α Feedforward Loop in Mouse Germline Stem Cells. Front Cell Dev Biol 2021; 9:646644. [PMID: 34381769 PMCID: PMC8351907 DOI: 10.3389/fcell.2021.646644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 06/03/2021] [Indexed: 01/16/2023] Open
Abstract
The mechanism on how extracellular matrix (ECM) cooperates with niche growth factors and oxygen tension to regulate the self-renewal of embryonic germline stem cells (GSCs) still remains unclear. Lacking of an appropriate in vitro cell model dramatically hinders the progress. Herein, using a serum-free culture system, we demonstrated that ECM laminin cooperated with hypoxia and insulin-like growth factor 1 receptor (IGF-1R) to additively maintain AP activity and Oct-4 expression of AP+GSCs. We found the laminin receptor CD49f expression in d2 testicular GSCs that were surrounded by laminin. Laminin and hypoxia significantly increased the GSC stemness-related genes, including Hif-2α, Oct-4, IGF-1R, and CD49f. Cotreatment of IGF-1 and laminin additively increased the expression of IGF-IR, CD49f, Hif-2α, and Oct-4. Conversely, silencing IGF-1R and/or CD49f decreased the expression of Hif-2α and Oct-4. The underlying mechanism involved CD49f/IGF1R-(PI3K/AKT)-Hif-2α signaling loop, which in turn maintains Oct-4 expression, symmetric self-renewal, and cell migration. These findings reveal the additive niche laminin/IGF-IR network during early GSC development.
Collapse
Affiliation(s)
- Heng-Kien Au
- Taipei Medical University (TMU) Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan.,Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.,International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Syue-Wei Peng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chin-Lin Guo
- Institute of Physics, Academia Sinica, Taipei, Taiwan
| | - Chien-Chia Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Lin Wang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yung-Che Kuo
- Taipei Medical University (TMU) Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tsz-Yau Law
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hong-Nerng Ho
- Taipei Medical University (TMU) Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, Taipei Municipal Wanfang Hospital, Taipei, Taiwan
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yen-Hua Huang
- Taipei Medical University (TMU) Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan.,Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.,International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Comprehensive Cancer Center of Taipei Medical University, Taipei, Taiwan.,The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
5
|
Ruan DT, Gao S, Shelat H, King B, Geng YJ. Differential expression of microRNA and arachidonic acid metabolism in aspirin-treated human cardiac and peri-cardiac fat-derived mesenchymal stem cells. Vascul Pharmacol 2020; 127:106651. [PMID: 32044414 DOI: 10.1016/j.vph.2020.106651] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 10/09/2019] [Accepted: 02/06/2020] [Indexed: 02/07/2023]
Abstract
Aspirin is a widely used drug with anti-coagulating and anti-inflammatory effects on atherosclerotic vascular disease. The goal of this study was to investigate expression of microRNA (miR) in association with changes in arachidonic acid (AA) metabolism in cardiac and surrounding fat mesenchymal stem cells (MSCs) treated with or without aspirin. Aspirin-targeted endogenous lipid metabolites that impact specific miRNA expression were examined by mass spectrometry. The pattern of miR expression was characterized using a microarray of 1100 miRs. There were a dozen miRs expressed differentially in MSCs from human myocardium and peri-myocardial fat tissue at baseline, including hsa-miR-1307-3p, 765, 4739, 3613-3p, 4281, 6816-5p, 2861, and 146b-5p. After exposure to aspirin, cardiac MSCs expressed an array of of miRs (eg, hsa-miR-4734, 10a-5p, 4267, 3197, and 3182), while generation of their endogenous AA metabolites was depressed. However, in the peri-cardiac adipose tissue-derived MSCs, treatment with the same doses of aspirin caused mild changes in the miR expression levels. In conclusion, MSCs from human myocardium and peri-myocardial fat tissue respond differentially to aspirin treatment by alterations in miR expression and AA metabolism. The study further raises an intriguing issue as to whether the copious amounts of aspirin taken worldwide by patients with cardiovascular disease may have direct impacts on their heart repair processes by regulation of stromal cell miR expression and AA metabolism.
Collapse
Affiliation(s)
- Diana T Ruan
- Columbia University Vagelos College of Physician and Surgeons, Columbia University, New York, NY, United States of America; Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Song Gao
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Harnath Shelat
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Bridgette King
- College of Agriculture and Life Science, Texas A&M University, College Station, TX, United States of America
| | - Yong-Jian Geng
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States of America.
| |
Collapse
|
6
|
Zhang W, Feng G, Wang L, Teng F, Wang L, Li W, Zhang Y, Zhou Q. MeCP2 deficiency promotes cell reprogramming by stimulating IGF1/AKT/mTOR signaling and activating ribosomal protein-mediated cell cycle gene translation. J Mol Cell Biol 2019; 10:515-526. [PMID: 29562294 DOI: 10.1093/jmcb/mjy018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 03/18/2018] [Indexed: 12/24/2022] Open
Abstract
The generation of induced pluripotent stem cells (iPSCs) offers a great opportunity in research and regenerative medicine. The current poor efficiency and incomplete mechanistic understanding of the reprogramming process hamper the clinical application of iPSCs. MeCP2 connects histone modification and DNA methylation, which are key changes of somatic cell reprogramming. However, the role of MeCP2 in cell reprogramming has not been examined. In this study, we found that MeCP2 deficiency enhanced reprogramming efficiency and stimulated cell proliferation through regulating cell cycle protein expression in the early stage of reprogramming. MeCP2 deficiency enhanced the expression of ribosomal protein genes, thereby enhancing reprogramming efficiency through promoting the translation of cell cycle genes. In the end, MeCP2 deficiency stimulated IGF1/AKT/mTOR signaling and activated ribosomal protein gene expression. Taken together, our data indicate that MeCP2 deficiency promoted cell reprogramming through stimulating IGF1/AKT/mTOR signaling and activating ribosomal protein-mediated cell cycle gene translation in the early stage of reprogramming.
Collapse
Affiliation(s)
- Wei Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Guihai Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Libin Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Fei Teng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Liu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
7
|
Bader R, Ibrahim JN, Moussa M, Mourad A, Azoury J, Azoury J, Alaaeddine N. In vitro
effect of autologous platelet‐rich plasma on H
2
O
2
‐induced oxidative stress in human spermatozoa. Andrology 2019; 8:191-200. [DOI: 10.1111/andr.12648] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/11/2019] [Accepted: 04/11/2019] [Indexed: 12/14/2022]
Affiliation(s)
- R. Bader
- Mount Lebanon HospitalAzoury IVF Clinic Beirut Lebanon
- Faculty of Medicine Regenerative Medicine and Inflammation Laboratory Saint‐Joseph University Beirut Lebanon
| | - J. N. Ibrahim
- Faculty of Public Health II Medical Laboratory Department Lebanese University Beirut Lebanon
| | - M. Moussa
- Faculty of Medicine Regenerative Medicine and Inflammation Laboratory Saint‐Joseph University Beirut Lebanon
| | - A. Mourad
- Department of Obstetrics and Gynecology American University of Beirut Medical Center Beirut Lebanon
| | - J. Azoury
- OB‐GYN Department Inova Fairfax Hospital Falls Church VA USA
| | - J. Azoury
- Mount Lebanon HospitalAzoury IVF Clinic Beirut Lebanon
| | - N. Alaaeddine
- Faculty of Medical Sciences Neuroscience Research CenterLebanese University Beirut Lebanon
| |
Collapse
|
8
|
Gupta MK, De Jesus DF, Kahraman S, Valdez IA, Shamsi F, Yi L, Swensen AC, Tseng YH, Qian WJ, Kulkarni RN. Insulin receptor-mediated signaling regulates pluripotency markers and lineage differentiation. Mol Metab 2018; 18:153-163. [PMID: 30316806 PMCID: PMC6308035 DOI: 10.1016/j.molmet.2018.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 09/07/2018] [Accepted: 09/11/2018] [Indexed: 12/30/2022] Open
Abstract
Objectives Insulin receptor (IR)-mediated signaling is involved in the regulation of pluripotent stem cells; however, its direct effects on regulating the maintenance of pluripotency and lineage development are not fully understood. The main objective of this study is to understand the role of IR signaling in pluripotency and lineage development. Methods To explore the role of IR signaling, we generated IR knock-out (IRKO) mouse induced pluripotent stem cells (miPSCs) from E14.5 mouse embryonic fibroblasts (MEFs) of global IRKO mice using a cocktail of four reprogramming factors: Oct4, Sox2, Klf4, cMyc. We performed pluripotency characterization and directed the differentiation of control and IRKO iPSCs into neural progenitors (ectoderm), adipocyte progenitors (mesoderm), and pancreatic beta-like cells (endoderm). We mechanistically confirmed these findings via phosphoproteomics analyses of control and IRKO iPSCs. Results Interestingly, expression of pluripotency markers including Klf4, Lin28a, Tbx3, and cMyc were upregulated, while abundance of Oct4 and Nanog were enhanced by 4-fold and 3-fold, respectively, in IRKO iPSCs. Analyses of signaling pathways demonstrated downregulation of phospho-STAT3, p-mTor and p-Erk and an increase in the total mTor and Erk proteins in IRKO iPSCs in the basal unstimulated state. Stimulation with leukemia inhibitory factor (LIF) showed a ∼33% decrease of phospho-ERK in IRKO iPSCs. On the contrary, Erk phosphorylation was increased during in vitro spontaneous differentiation of iPSCs lacking IRs. Lineage-specific directed differentiation of the iPSCs revealed that cells lacking IR showed enhanced expression of neuronal lineage markers (Pax6, Tubb3, Ascl1 and Oligo2) while exhibiting a decrease in adipocyte (Fas, Acc, Pparγ, Fabp4, C/ebpα, and Fsp27) and pancreatic beta cell markers (Ngn3, Isl1, and Sox9). Further molecular characterization by phosphoproteomics confirmed the novel IR-mediated regulation of the global pluripotency network including several key proteins involved in diverse aspects of growth and embryonic development. Conclusion We report, for the first time to our knowledge, the phosphoproteome of insulin, IGF1, and LIF stimulation in mouse iPSCs to reveal the importance of insulin receptor signaling for the maintenance of pluripotency and lineage determination. Insulin receptor signaling regulates expression of key pluripotency genes including Oct4 and Nanog. IRKO iPSCs show upregulation of neuronal markers during differentiation. Adipocyte and pancreatic beta cell differentiation are perturbed in IRKO iPSCs. Phosphoproteomics analyses confirmed the role of IR in regulation of pluripotency and developmental proteins.
Collapse
Affiliation(s)
- Manoj K Gupta
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Dario F De Jesus
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA; Graduate Program in Areas of Basic and Applied Biology (GABBA), Abel Salazar Biomedical Sciences Institute, University of Porto, 5000 Porto, Portugal
| | - Sevim Kahraman
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Ivan A Valdez
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Farnaz Shamsi
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lian Yi
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Adam C Swensen
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Yu-Hua Tseng
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
9
|
An active IGF-1R-AKT signaling imparts functional heterogeneity in ovarian CSC population. Sci Rep 2016; 6:36612. [PMID: 27819360 PMCID: PMC5098199 DOI: 10.1038/srep36612] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/17/2016] [Indexed: 02/06/2023] Open
Abstract
Deregulated IGF-1R-AKT signaling influences multiple nodes of cancer cell physiology and assists in migration, metastasis and acquirement of radio/chemoresistance. Enrichment of cancer stem cells (CSC) positively correlates with radio/chemoresistance development in various malignancies. It is unclear though, how IGF-1R-AKT signalling shapes CSC functionality especially in ovarian cancer. Previously we showed that upregulated IGF-1R expression is essential to initiate platinum-taxol resistance at early stage which declines with elevated levels of activated AKT at late resistant stage in ovarian cancer cells. Here, we investigated the effect of this oscillatory IGF-1R-AKT signalling upon CSC functionality during generation of chemoresistance. While gradual increase in CSC properties from early (ER) to late (LR) resistant stages was observed in three different (cisplatin/paclitaxel/cisplatin-paclitaxel) cellular models created in two ovarian cancer cell lines, the stemness gene expressions (oct4/sox2/nanog) reached a plateau at early resistant stages. Inhibition of IGF-1R only at ER and AKT inhibition only at LR stages significantly abrogated the CSC phenotype. Interestingly, real time bioluminescence imaging showed CSCs of ER stages possessed faster tumorigenic potential than CSCs belonging to LR stages. Together, our data suggest that IGF-1R-AKT signalling imparts functional heterogeneity in CSCs during acquirement of chemoresistance in ovarian carcinoma.
Collapse
|
10
|
Kang X, Wei X, Jiang L, Niu C, Zhang J, Chen S, Meng D. Nox2 and Nox4 regulate self-renewal of murine induced-pluripotent stem cells. IUBMB Life 2016; 68:963-970. [PMID: 27797149 DOI: 10.1002/iub.1574] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/19/2016] [Indexed: 12/15/2022]
Abstract
Reactive oxygen species (ROS) and redox homeostasis have a pivotal role in the maintenance of stem cell pluripotency and in stem cell self-renewal; however, the mechanisms by which ROS regulate the self-renewal of stem cells have not been thoroughly studied. Here, we evaluated the role of the ROS produced by NADPH oxidase 2 (Nox2) and NADPH oxidase 4 (Nox4) in the self-renewal and stemness of murine induced-pluripotent stem cells (miPSCs). Targeted silencing of Nox2 or Nox4 reduced both NADPH oxidase activity and intracellular ROS levels, as well as alkaline phosphatase activity, the total number of miPSCs, the expression of insulin-like growth factor-1 (IGF-1), IGF-1 receptor, and the phosphorylation of extracellular signal regulated kinase (ERK) 1/2. Nox2/Nox4 overexpression or low, nontoxic concentration of H2 O2 increased cell proliferation in miPSCs. Furthermore, expression of the stemness genes Sox2 and Oct4 was lower in Nox2/Nox4-deficient miPSCs, and higher in Nox2/Nox4-overexpressing miPSCs, than in miPSCs with normal levels of Nox2/Nox4 expression. Collectively, these results suggest that Nox2- and Nox4-derived ROS contribute to stem cell pluripotency maintenance and self-renewal. © 2016 IUBMB Life, 68(12):963-970, 2016.
Collapse
Affiliation(s)
- Xueling Kang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiangxiang Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Li Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Cong Niu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Sifeng Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Dan Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Wang J, Cao W, Niu F. Adenoviral vector expressing IGF-1 protects murine chondrogenic ATDC5 cells against hydrogen peroxide-induced mitochondrial dysfunction and apoptosis. J Toxicol Sci 2016; 40:585-95. [PMID: 26354375 DOI: 10.2131/jts.40.585] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Insulin-like growth factor-1 (IGF-1), with an age-related decline, regulates the proliferation and survival of multiple cell types, particularly stimulates cartilage matrix synthesis, and inhibits matrix degradation. The present study was to investigate the regulatory role of IGF-1 against hydrogen peroxide(H2O2)-induced mitochondrial dysfunction and apoptosis in murine chondrocytic ATDC5 cells. We firstly determined mitochondrial dysfunction and apoptosis in ATDC5 cells which were exposed to H2O2. We then constructed an IGF-1-overexpressed adenovirus (IGF-1-Ad) harboring the IGF-1 coding sequence, and investigated the regulatory role of the overexpressed IGF-1 against the H2O2-induced mitochondrial dysfunction and apoptosis in ATDC5 cells. It was demonstrated that H2O2 treatment promoted the mitochondrial dysfunction, and further reduced the viability and induced apoptosis of ATDC5 cells. However, the IGF-1 overexpression by adenovirus inhibited the H2O2-induced mitochondrial dysfunction and further inhibited the H2O2-promoted apoptosis in ATDC5 cells. In conclusion, the present study found that oxidative stress promoted mitochondrial dysfunction and induced apoptosis in the murine chondrocytic ATDC5 cells, and the adenoviral vector-expressed IGF-1 protected the murine chondrocytic ATDC5 cells against such mitochondrial dysfunction and apoptosis. This study implies the protective role of IGF-1 against the oxidative stress in murine chondrocytic ATDC5 cells and demonstrates the promising anti-oxidative stress effect of the recombinant IGF-1-Ad against oxidative stress in chondrocytic cells.
Collapse
Affiliation(s)
- Jinfeng Wang
- Department of Orthopaedics, Tianjin 4th Centre Hospital, China
| | | | | |
Collapse
|
12
|
Youssef A, Han VKM. Low Oxygen Tension Modulates the Insulin-Like Growth Factor-1 or -2 Signaling via Both Insulin-Like Growth Factor-1 Receptor and Insulin Receptor to Maintain Stem Cell Identity in Placental Mesenchymal Stem Cells. Endocrinology 2016; 157:1163-74. [PMID: 26760116 DOI: 10.1210/en.2015-1297] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Placental mesenchymal stem cells (PMSCs) are readily available multipotent stem cells for potential use in regenerative therapies. For this purpose, PMSCs must be maintained in culture conditions that mimic the in vivo microenvironment. IGFs (IGF-1 and IGF-2) and oxygen tension are low in the placenta in early gestation and increase as pregnancy progresses. IGFs bind to two receptor tyrosine kinases, the IGF-1 receptor (IGF-1R) and the insulin receptor (IR), and their hybrid receptors. We hypothesized that IGF-1 and IGF-2 signal via distinct signaling pathways under low-oxygen tension to maintain PMSC multipotency. In preterm PMSCs, low-oxygen tension increased the expression of IGF-2 and reduced IGF-1. IGF-1 stimulated higher phosphorylation of IGF-1Rβ, ERK1/2, and AKT, which was maintained at steady lower levels by low oxygen tension. PMSC proliferation was increased by IGF-1 more than IGF-2,and was potentiated by low-oxygen tension. This IGF/low oxygen tension-mediated proliferation was receptor dependent because neutralization of the IGF-1R inhibited PMSC proliferation in the presence of IGF-1 and the IR in presence of IGF-2. These findings suggest that both IGF-1R and the IR can participate in mediating IGF signaling in maintaining PMSCs multipotency. We conclude that low-oxygen tension can modify the IGF-1 or IGF-2 signaling via the IGF-1R and IR in PMSCs.
Collapse
Affiliation(s)
- Amer Youssef
- Departments of Biochemistry (A.Y., V.K.M.H.) and Paediatrics (V.K.M.H.), Schulich School of Medicine and Dentistry, Children's Health Research Institute (A.Y., V.K.M.H.), and Lawson Health Research Institute (A.Y., V.K.M.H.), Western University, London, Ontario, Canada N6C 2V5
| | - Victor K M Han
- Departments of Biochemistry (A.Y., V.K.M.H.) and Paediatrics (V.K.M.H.), Schulich School of Medicine and Dentistry, Children's Health Research Institute (A.Y., V.K.M.H.), and Lawson Health Research Institute (A.Y., V.K.M.H.), Western University, London, Ontario, Canada N6C 2V5
| |
Collapse
|
13
|
Abstract
Neural stem cells (NSCs) are found in two regions in the adult brain: the subgranular zone (SGZ) in the hippocampal dentate gyrus and the subventricular zone (SVZ) adjacent to the lateral ventricles. Similarly to other somatic stem cells, adult NSCs are found within specialized niches that are organized to facilitate NSC self-renewal. Alterations in stem-cell homeostasis can contribute to the consequences of neurodegenerative diseases, healthy ageing and tissue repair after damage. Insulin and the insulin-like growth factors (IGFs) function in stem-cell homeostasis across species. Studies in the mammalian central nervous system support essential roles for IGF and/or insulin signalling in NSC self-renewal, neurogenesis, cognition and sensory function through distinct ligand-receptor interactions. IGF-II is of particular interest as a result of its production by the choroid plexus and presence in cerebrospinal fluid (CSF). CSF regulates and supports the development, division and migration of cells in the adult brain and is required for NSC maintenance. In this Review, we discuss emerging data on the functions of IGF-II and IGF and/or insulin receptor signalling in the context of NSC regulation in the SVZ and SGZ. We also propose a model for IGF-II in which the choroid plexus is a major component of the NSC niche.
Collapse
Affiliation(s)
- Amber N Ziegler
- Department of Neurology &Neuroscience, New Jersey Medical School, Rutgers Biomedical &Health Sciences, Cancer Centre, 205 South Orange Avenue, Newark, NJ 07101, USA
| | - Steven W Levison
- Department of Neurology &Neuroscience, New Jersey Medical School, Rutgers Biomedical &Health Sciences, Cancer Centre, 205 South Orange Avenue, Newark, NJ 07101, USA
| | - Teresa L Wood
- Department of Neurology &Neuroscience, New Jersey Medical School, Rutgers Biomedical &Health Sciences, Cancer Centre, 205 South Orange Avenue, Newark, NJ 07101, USA
| |
Collapse
|
14
|
Ramadhin C, Pillay B, Olaniran AO. Cell-based assays for IGF-I bioactivity measurement: overview, limitations and current trends. Growth Factors 2014; 32:130-8. [PMID: 25060037 DOI: 10.3109/08977194.2014.939806] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Insulin-like growth factor-I (IGF-I) is an important growth promoting protein that is involved in numerous cellular responses and multiple biological systems. Although the molecular structure, function and recombinant production of IGF-I in various hosts have been the subject of much researches over the recent past, methods to determine the bioactivity of this protein have not been fully explored. Several assays have traditionally been used to measure IGF-I bioactivity, but have not become a routine laboratory practice due to the high cost involved and technical problems. Thus, there is still a need for a rapid, technically simple and accurate assay to determine IGF-I bioactivity. This review highlights the various cell-based assays currently commercially available for measuring the bioactivity of IGF-I along with their limitations. This is aimed at presenting the modern-day IGF researcher with a holistic overview of the current trends and future prospects regarding IGF-I bioactivity determinations.
Collapse
Affiliation(s)
- Charlotte Ramadhin
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal , Durban , Republic of South Africa
| | | | | |
Collapse
|
15
|
Huang YH, Lin MH, Wang PC, Wu YC, Chiang HL, Wang YL, Chang JH, Huang YK, Gu SY, Ho HN, Ling TY. Hypoxia inducible factor 2α/insulin-like growth factor receptor signal loop supports the proliferation and Oct-4 maintenance of mouse germline stem cells. Mol Hum Reprod 2014; 20:526-37. [PMID: 24598112 DOI: 10.1093/molehr/gau016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Hypoxia inducible factor 2α (HIF-2α) is critical for primordial germ cell (PGC) survival as knockout of HIF-2α (HIF-2α(-/-)) decreases both expression of Oct-4 and PGC number in genital ridge. Hypoxia is known to stabilize HIF-2α protein from proteasomal degradation. However, little is known about the hypoxia-associated endocrinal signaling in HIF-2α expression. The current work demonstrates a role for an endocrine insulin-like growth factor-I receptor (IGF-IR)-PI3K/Akt-mTOR-HIF-2α regulatory loop in the proliferation and Oct-4 maintenance of PGC-like alkaline phosphatase positive mouse germline stem cells (AP(+)GSCs). We found that hypoxia greatly increased the cell proliferation and the levels of nuclear Oct-4/HIF-2α protein of AP(+)GSCs. The hypoxic-AP(+)GSCs presented stronger stemness ability for germ cell differentiation than normoxic, with expressions of c-KIT (differentiation germ cell marker), VASA (differentiation germ cell marker) and SCP3 (meiotic marker) using a renal capsule transplantation assay. Meanwhile, hypoxia significantly increased the expression levels of secreted-IGF-I and IGF-IR. The IGF-I dose dependently increased the HIF-2α expression levels in AP(+)GSCs; and, the inhibition of IGF-IR by RNA interference (shIGF-IR) or LY294002 (PI3K inhibitor)/Rapamycin (mTOR inhibitor) effectively suppressed the IGF-I- and/or hypoxia-induced HIF-2α and Oct-4 expression, suggesting that the IGF-IR and its downstream Akt/mTOR signaling are involved in the IGF-I/hypoxia effects. Additionally, knockdown of HIF-2α dramatically suppressed Oct-4 and IGF-IR protein levels in AP(+)GSC cells. In conclusion, the present study demonstrates a regulatory loop of IGF-IR-PI3K/Akt-mTOR-HIF-2α in proliferation and Oct-4 maintenance of PGC-like AP(+)GSCs under hypoxia. This finding provides insights into the niche endocrinology underlying early germ cell development.
Collapse
Affiliation(s)
- Y-H Huang
- Department of Biochemistry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan Center for Reproductive Medicine and Sciences, Taipei Medical University Hospital, Taipei, Taiwan
| | - M-H Lin
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - P-C Wang
- Department of Biochemistry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Y-C Wu
- Department of Biochemistry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - H-L Chiang
- Department of Biochemistry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Y-L Wang
- Department of Biochemistry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - J-H Chang
- Department of Biochemistry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Y-K Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, No.1, Section 1, Je-Ai Rd, Taipei, Taiwan
| | - S-Y Gu
- Department of Pharmacology, College of Medicine, National Taiwan University, No.1, Section 1, Je-Ai Rd, Taipei, Taiwan
| | - H-N Ho
- Graduate Institute of Clinical Genomics, College of Medicine, National Taiwan University, Taipei, Taiwan Division of Endocrinology and Infertility, Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei Medical University, Taipei, Taiwan
| | - T-Y Ling
- Department of Pharmacology, College of Medicine, National Taiwan University, No.1, Section 1, Je-Ai Rd, Taipei, Taiwan
| |
Collapse
|
16
|
Vandomme J, Touil Y, Ostyn P, Olejnik C, Flamenco P, El Machhour R, Segard P, Masselot B, Bailliez Y, Formstecher P, Polakowska R. Insulin-like growth factor 1 receptor and p38 mitogen-activated protein kinase signals inversely regulate signal transducer and activator of transcription 3 activity to control human dental pulp stem cell quiescence, propagation, and differentiation. Stem Cells Dev 2014; 23:839-51. [PMID: 24266654 DOI: 10.1089/scd.2013.0400] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Dental pulp stem cells (DPSCs) remain quiescent until activated in response to severe dental pulp damage. Once activated, they exit quiescence and enter regenerative odontogenesis, producing reparative dentin. The factors and signaling molecules that control the quiescence/activation and commitment to differentiation of human DPSCs are not known. In this study, we determined that the inhibition of insulin-like growth factor 1 receptor (IGF-1R) and p38 mitogen-activated protein kinase (p38 MAPK) signaling commonly activates DPSCs and promotes their exit from the G0 phase of the cell cycle as well as from the pyronin Y(low) stem cell compartment. The inhibition of these two pathways, however, inversely determines DPSC fate. In contrast to p38 MAPK inhibitors, IGF-1R inhibitors enhance dental pulp cell sphere-forming capacity and reduce the cells' colony-forming capacity without inducing cell death. The inverse cellular changes initiated by IGF-1R and p38 MAPK inhibitors were accompanied by inverse changes in the levels of active signal transducer and activator of transcription 3 (STAT3) factor, inactive glycogen synthase kinase 3, and matrix extracellular phosphoglycoprotein, a marker of early odontoblast differentiation. Our data suggest that there is cross talk between the IGF-1R and p38 MAPK signaling pathways in DPSCs and that the signals provided by these pathways converge at STAT3 and inversely regulate its activity to maintain quiescence or to promote self-renewal and differentiation of the cells. We propose a working model that explains the possible interactions between IGF-1R and p38 MAPK at the molecular level and describes the cellular consequences of these interactions. This model may inspire further fundamental study and stimulate research on the clinical applications of DPSC in cellular therapy and tissue regeneration.
Collapse
Affiliation(s)
- Jerome Vandomme
- 1 Inserm U837 Jean-Pierre Aubert Research Center, Institut pour la Recherche sur le Cancer de Lille (IRCL) , Lille, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Castaño Z, Marsh T, Tadipatri R, Kuznetsov HS, Al-Shahrour F, Paktinat M, Greene-Colozzi A, Nilsson B, Richardson AL, McAllister SS. Stromal EGF and igf-I together modulate plasticity of disseminated triple-negative breast tumors. Cancer Discov 2013; 3:922-35. [PMID: 23689072 DOI: 10.1158/2159-8290.cd-13-0041] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The causes for malignant progression of disseminated tumors and the reasons recurrence rates differ in women with different breast cancer subtypes are unknown. Here, we report novel mechanisms of tumor plasticity that are mandated by microenvironmental factors and show that recurrence rates are not strictly due to cell-intrinsic properties. Specifically, outgrowth of the same population of incipient tumors is accelerated in mice with triple-negative breast cancer (TNBC) relative to those with luminal breast cancer. Systemic signals provided by overt TNBCs cause the formation of a tumor-supportive microenvironment enriched for EGF and insulin-like growth factor-I (IGF-I) at distant indolent tumor sites. Bioavailability of EGF and IGF-I enhances the expression of transcription factors associated with pluripotency, proliferation, and epithelial-mesenchymal transition. Combinatorial therapy with EGF receptor and IGF-I receptor inhibitors prevents malignant progression. These results suggest that plasticity and recurrence rates can be dictated by host systemic factors and offer novel therapeutic potential for patients with TNBC.
Collapse
Affiliation(s)
- Zafira Castaño
- Hematology Division , Brigham & Women's Hospital, Lund University, Lund, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Xu C, Xie D, Yu SC, Yang XJ, He LR, Yang J, Ping YF, Wang B, Yang L, Xu SL, Cui W, Wang QL, Fu WJ, Liu Q, Qian C, Cui YH, Rich JN, Kung HF, Zhang X, Bian XW. β-Catenin/POU5F1/SOX2 transcription factor complex mediates IGF-I receptor signaling and predicts poor prognosis in lung adenocarcinoma. Cancer Res 2013; 73:3181-9. [PMID: 23539445 DOI: 10.1158/0008-5472.can-12-4403] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer stem-like cells (CSLC) are crucial in tumor initiation and progression; however, the underlying mechanism for the self-renewal of cancer cells remains undefined. In the study, immunohistochemical analysis of specimens freshly excised from patients with lung adenocarcinoma showed that high expression of insulin-like growth factor I receptor (IGF-IR) in lung adenocarcinoma cells was positively correlated with the expressions of cancer stem cell markers CD133 and aldehyde dehydrogenase 1 family member A1 (ALDH1A1). IGF-IR activation enhanced POU class 5 homeobox 1 (POU5F1) expression on human lung adenocarcinoma stem-like cells (LACSLC) through PI3K/AKT/GSK3β/β-catenin cascade. POU5F1 could form a novel complex with β-catenin and SOX2 to bind Nanog promoter for transcription to maintain self-renewal of LACSLCs, which was dependent on the functional IGF-IR. Genetic and pharmacologic inhibition of IGF-IR abrogated LACSLC capabilities for self-renewal and tumorigenicity in vitro. In an in vivo xenograft tumor model, knockdown of either IGF-IR or POU5F1 impeded tumorigenic potentials of LACSLCs. By analyzing pathologic specimens excised from 200 patients with lung adenocarcinoma, we found that colocalization of highly expressed IGF-IR with β-catenin and POU5F1 predicted poor prognosis. Taken together, we show that IGF-IR-mediated POU5F1 expression to form a complex with β-catenin and SOX2 is crucial for the self-renewal and oncogenic potentials of LACSLCs, and the integrative clinical detection of the expressions of IGF-IR, β-catenin, and POU5F1 is indicatory for predicting prognosis in the patients of lung adenocarcinoma.
Collapse
Affiliation(s)
- Chuan Xu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Ziegler AN, Schneider JS, Qin M, Tyler WA, Pintar JE, Fraidenraich D, Wood TL, Levison SW. IGF-II promotes stemness of neural restricted precursors. Stem Cells 2012; 30:1265-76. [PMID: 22593020 DOI: 10.1002/stem.1095] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Insulin-like growth factor (IGF)-I and IGF-II regulate brain development and growth through the IGF type 1 receptor (IGF-1R). Less appreciated is that IGF-II, but not IGF-I, activates a splice variant of the insulin receptor (IR) known as IR-A. We hypothesized that IGF-II exerts distinct effects from IGF-I on neural stem/progenitor cells (NSPs) via its interaction with IR-A. Immunofluorescence revealed high IGF-II in the medial region of the subventricular zone (SVZ) comprising the neural stem cell niche, with IGF-II mRNA predominant in the adjacent choroid plexus. The IGF-1R and the IR isoforms were differentially expressed with IR-A predominant in the medial SVZ, whereas the IGF-1R was more abundant laterally. Similarly, IR-A was more highly expressed by NSPs, whereas the IGF-1R was more highly expressed by lineage restricted cells. In vitro, IGF-II was more potent in promoting NSP expansion than either IGF-I or standard growth medium. Limiting dilution and differentiation assays revealed that IGF-II was superior to IGF-I in promoting stemness. In vivo, NSPs propagated in IGF-II migrated to and took up residence in periventricular niches while IGF-I-treated NSPs predominantly colonized white matter. Knockdown of IR or IGF-1R using shRNAs supported the conclusion that the IGF-1R promotes progenitor proliferation, whereas the IR is important for self-renewal. Q-PCR revealed that IGF-II increased Oct4, Sox1, and FABP7 mRNA levels in NSPs. Our data support the conclusion that IGF-II promotes the self-renewal of neural stem/progenitors via the IR. By contrast, IGF-1R functions as a mitogenic receptor to increase precursor abundance.
Collapse
Affiliation(s)
- Amber N Ziegler
- Department of Neurology and Neuroscience, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, New Jersey, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Li Y, Shelat H, Geng YJ. IGF-1 prevents oxidative stress induced-apoptosis in induced pluripotent stem cells which is mediated by microRNA-1. Biochem Biophys Res Commun 2012; 426:615-9. [PMID: 22982320 DOI: 10.1016/j.bbrc.2012.08.139] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 08/29/2012] [Indexed: 11/18/2022]
Abstract
Oxidative stress contributes to tissue injury and cell death during the development of various diseases. The present study aims at investigating whether oxidative stress triggered by the exposure to hydrogen peroxide (H(2)O(2)) can induce apoptosis of induced pluripotent stem cells (iPS cells) in a mechanism mediated by insulin-like growth factor (IGF-1) and microRNA-1 (miR-1). iPS cells treated with H(2)O(2) showed increases in miR-1 expression, mitochondria dysfunction, cytochrome-c release and apoptosis, Addition of IGF-1 into the iPS cell cultures reduced the H(2)O(2) cytotoxicity. Prediction algorithms showed that 3'-untranslated regions of IGF-1 gene as a target of miR-1. Moreover, miR-1 mimic, but not miR-1 mimic negative control, diminished the protective effect of IGF-1 on H(2)O(2)-induced mitochondrial dysfunction, cytochrome-c release and apoptosis in iPS cells. In conclusion, IGF-1 inhibits H(2)O(2)-induced mitochondrial dysfunction, cytochrome-c release and apoptosis. IGF-1's effect is, at least partially, regulated by miR-1 in iPS cells.
Collapse
Affiliation(s)
- Yangxin Li
- The Texas Heart Institute and Center for Cardiovascular and Atherosclerosis Research, Department of Internal Medicine, University of Texas Medical School at Houston, TX 77030, USA.
| | | | | |
Collapse
|
21
|
Shan J, Shen J, Liu L, Xia F, Xu C, Duan G, Xu Y, Ma Q, Yang Z, Zhang Q, Ma L, Liu J, Xu S, Yan X, Bie P, Cui Y, Bian XW, Qian C. Nanog regulates self-renewal of cancer stem cells through the insulin-like growth factor pathway in human hepatocellular carcinoma. Hepatology 2012; 56:1004-14. [PMID: 22473773 DOI: 10.1002/hep.25745] [Citation(s) in RCA: 249] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Accepted: 03/19/2012] [Indexed: 12/11/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) exhibits cellular heterogeneity and embryonic stem-cell-related genes are preferentially overexpressed in a fraction of cancer cells of poorly differentiated tumors. However, it is not known whether or how these cancer cells contribute to tumor initiation and progression. Here, our data showed that increased expression of pluripotency transcription factor Nanog in cancer cells correlates with a worse clinical outcome in HCC. Using the Nanog promoter as a reporter system, we could successfully isolate a small subpopulation of Nanog-positive cells. We demonstrate that Nanog-positive cells exhibited enhanced ability of self-renewal, clonogenicity, and initiation of tumors, which are consistent with crucial hallmarks in the definition of cancer stem cells (CSCs). Nanog(Pos) CSCs could differentiate into mature cancer cells in in vitro and in vivo conditions. In addition, we found that Nanog(Pos) CSCs exhibited resistance to therapeutic agents (e.g., sorafenib and cisplatin) and have a high capacity for tumor invasion and metastasis. Knock-down expression of Nanog in Nanog(Pos) CSCs could decrease self-renewal accompanied with decreased expression of stem-cell-related genes and increased expression of mature hepatocyte-related genes. Overexpression of Nanog in Nanog(Neg) cells could restore self-renewal. Furthermore, we found that insulin-like growth factor (IGF)2 and IGF receptor (IGF1R) were up-regulated in Nanog(Pos) CSCs. Knock-down expression of Nanog in Nanog(Pos) CSCs inhibited the expression of IGF1R, and overexpression of Nanog in Nanog(Neg) cells increased the expression of IGF1R. A specific inhibitor of IGF1R signaling could significantly inhibit self-renewal and Nanog expression, indicating that IGF1R signaling participated in Nanog-mediated self-renewal. CONCLUSION These data indicate that Nanog could be a novel biomarker for CSCs in HCC, and that Nanog could play a crucial role in maintaining the self-renewal of CSCs through the IGF1R-signaling pathway.
Collapse
Affiliation(s)
- Juanjuan Shan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
The bed and the bugs: interactions between the tumor microenvironment and cancer stem cells. Semin Cancer Biol 2012; 22:462-70. [PMID: 22548722 DOI: 10.1016/j.semcancer.2012.04.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 04/16/2012] [Indexed: 12/26/2022]
Abstract
Tumors have been increasingly recognized as organs with a complexity that approaches, and may even exceed, that of healthy tissues. When viewed from this perspective, the biology of a tumor can be understood only by studying tumor cell heterogeneity and the microenvironment that is constructed during the course of tumorigenesis and malignant progression. Recent work has revealed the existence of cancer stem cells, the "bugs", with the capacity for self-renewal and tumor propagation. In addition, it is now recognized that the tumor microenvironment, the "bed", plays a critical role in supporting cancer stem cells and also may promote neoplasia and malignant progression. The interdependence of the cell-intrinsic features of cancer, including the cancer stem cell "bugs" and the tumor microenvironment "bed", is only beginning to be understood. In this review, we highlight the rapidly evolving concepts about the interactions between tumor stem cells and their microenvironment, the insights gained from studying their normal tissue counterparts, and the questions and controversies surrounding this area of research, with an emphasis on breast and lung cancer. Finally, we address evidence supporting the notion that eliminating the bed as well as the bugs should lead to more effective and personalized cancer treatments that improve patient outcome.
Collapse
|
23
|
Lopez-Dee ZP, Chittur SV, Patel B, Stanton R, Wakeley M, Lippert B, Menaker A, Eiche B, Terry R, Gutierrez LS. Thrombospondin-1 type 1 repeats in a model of inflammatory bowel disease: transcript profile and therapeutic effects. PLoS One 2012; 7:e34590. [PMID: 22509329 PMCID: PMC3318003 DOI: 10.1371/journal.pone.0034590] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 03/02/2012] [Indexed: 12/31/2022] Open
Abstract
Thrombospondin-1 (TSP-1) is a matricellular protein with regulatory functions in inflammation and cancer. The type 1 repeats (TSR) domains of TSP-1 have been shown to interact with a wide range of proteins that result in the anti-angiogenic and anti-tumor properties of TSP-1. To ascertain possible functions and evaluate potential therapeutic effects of TSRs in inflammatory bowel disease, we conducted clinical, histological and microarray analyses on a mouse model of induced colitis. We used dextran sulfate sodium (DSS) to induce colitis in wild-type (WT) mice for 7 days. Simultaneously, mice were injected with either saline or one form of TSP-1 derived recombinant proteins, containing either (1) the three type 1 repeats of the TSP-1 (3TSR), (2) the second type 1 repeat (TSR2), or (3) TSR2 with the RFK sequence (TSR2+RFK). Total RNA isolated from the mice colons were processed and hybridized to mouse arrays. Array data were validated by real-time qPCR and immunohistochemistry. Histological and disease indices reveal that the mice treated with the TSRs show different patterns of leukocytic infiltration and that 3TSR treatment was the most effective in decreasing inflammation in DSS-induced colitis. Transcriptional profiling revealed differentially expressed (DE) genes, with the 3TSR-treated mice showing the least deviation from the WT-water controls. In conclusion, this study shows that 3TSR treatment is effective in attenuating the inflammatory response to DSS injury. In addition, the transcriptomics work unveils novel genetic data that suggest beneficial application of the TSR domains in inflammatory bowel disease.
Collapse
Affiliation(s)
- Zenaida P. Lopez-Dee
- Department of Biology, Wilkes University, Wilkes-Barre, Pennsylvania, United States of America
| | - Sridar V. Chittur
- Center for Functional Genomics, University at Albany, State University of New York, Rensselaer, New York, United States of America
| | - Bhumi Patel
- Department of Biology, Wilkes University, Wilkes-Barre, Pennsylvania, United States of America
| | - Rebecca Stanton
- Department of Biology, Wilkes University, Wilkes-Barre, Pennsylvania, United States of America
| | - Michelle Wakeley
- Department of Biology, Wilkes University, Wilkes-Barre, Pennsylvania, United States of America
| | - Brittany Lippert
- Department of Biology, Wilkes University, Wilkes-Barre, Pennsylvania, United States of America
| | - Anastasya Menaker
- Department of Biology, Wilkes University, Wilkes-Barre, Pennsylvania, United States of America
| | - Bethany Eiche
- Department of Biology, Wilkes University, Wilkes-Barre, Pennsylvania, United States of America
| | - Robert Terry
- Department of Biology, Wilkes University, Wilkes-Barre, Pennsylvania, United States of America
| | - Linda S. Gutierrez
- Department of Biology, Wilkes University, Wilkes-Barre, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
24
|
Pistollato F, Bremer-Hoffmann S, Healy L, Young L, Stacey G. Standardization of pluripotent stem cell cultures for toxicity testing. Expert Opin Drug Metab Toxicol 2012; 8:239-57. [PMID: 22248265 DOI: 10.1517/17425255.2012.639763] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Pluripotent stem cell (PSC) lines offer a unique opportunity to derive various human cell types that can be exploited for human safety assessments in vitro and as such contribute to modern mechanistically oriented toxicity testing. AREAS COVERED This article reviews the two major types of PSC cultures that are currently most promising for toxicological applications: human embryonic stem cell lines and human induced PSC lines. Through the review, the article explains how these cell types will improve the current safety evaluations of chemicals and will allow a more efficient selection of drug candidates. Additionally, the article discusses the important issues of maintaining PSCs as well as their differentiation efficiency. EXPERT OPINION The demonstration of the reliability and relevance of in vitro toxicity tests for a given purpose is mandatory for their use in regulatory toxicity testing. Given the peculiar nature of PSCs, a high level of standardization of undifferentiated cell cultures as well as of the differentiation process is required in order to ensure the establishment of robust test systems. It is, therefore, of pivotal importance to define and internationally agree on crucial parameters to judge the quality of the cellular models before enrolling them for toxicity testing.
Collapse
Affiliation(s)
- Francesca Pistollato
- Institute for Health & Consumer Protection, Systems Toxicology Unit, Joint Research Centre, European Commission, Ispra, Italy
| | | | | | | | | |
Collapse
|
25
|
Zhu L, del Vecchio G, de Micheli G, Liu Y, Carrara S, Calzà L, Nardini C. Biochips for Regenerative Medicine: Real-time Stem Cell Continuous Monitoring as Inferred by High-Throughput Gene Analysis. BIONANOSCIENCE 2011. [DOI: 10.1007/s12668-011-0028-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
26
|
Zhao Y, Brezina P, Hsu CC, Garcia J, Brinsden PR, Wallach E. In vitro fertilization: Four decades of reflections and promises. Biochim Biophys Acta Gen Subj 2011; 1810:843-52. [DOI: 10.1016/j.bbagen.2011.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 05/03/2011] [Accepted: 05/04/2011] [Indexed: 12/30/2022]
|