51
|
Zhang Z, Huang C, Wang P, Gao J, Liu X, Li Y, Yan S, Shi Y. HIF‑1α affects trophoblastic apoptosis involved in the onset of preeclampsia by regulating FOXO3a under hypoxic conditions. Mol Med Rep 2020; 21:2484-2492. [PMID: 32323858 PMCID: PMC7185296 DOI: 10.3892/mmr.2020.11050] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 02/07/2020] [Indexed: 12/25/2022] Open
Abstract
Preeclampsia (PE) is a pregnancy-specific syndrome that has severe implications on perinatal mortality and morbidity. Excessive apoptosis of trophoblasts induced by hypoxia may be associated with the development of PE, but the exact pathogenesis is unknown. Forkhead box O transcription factor 3a (FOXO3a) is activated under hypoxic conditions. Furthermore, hypoxia-inducible factor-1α (HIF-1α) is sensitive to variations in partial oxygen pressure. Thus, the aims of the present study were to investigate the expression levels of HIF-1α and FOXO3a in placental samples of early onset severe PE, and their effect on trophoblastic apoptosis under hypoxic conditions. Cobalt chloride was used to establish the hypoxic model. The present study examined the expression levels of HIF-1α and FOXO3a in the placental tissues and HTR8/SVneo cells under hypoxic conditions. It was found that HIF-1α and FOXO3a were highly expressed in placental tissues of patients with PE and in HTR8/SVneo cells under hypoxic conditions. Furthermore, knockdown of FOXO3a using a specific small interfering RNA (siRNA) decreased apoptosis in HTR8/SVneo cells. Moreover, it was found that after knockdown of HIF-1α using siRNA, FOXO3a expression and the apoptotic rate were reduced in HTR8/SVneo cells. Therefore, the present results indicated that the elevated expression of HIF-1α increased trophoblastic apoptosis by regulating FOXO3a, which may be involved in the pathogenesis of PE.
Collapse
Affiliation(s)
- Zhan Zhang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chenxi Huang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Ping Wang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Junjun Gao
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xin Liu
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yingying Li
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shujun Yan
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Ying Shi
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
52
|
Watanabe N, Morimatsu M, Fujita A, Teranishi M, Sudevan S, Watanabe M, Iwasa H, Hata Y, Kagi H, Nishiyama M, Naruse K, Higashitani A. Increased hydrostatic pressure induces nuclear translocation of DAF-16/FOXO in C. elegans. Biochem Biophys Res Commun 2020; 523:853-858. [PMID: 31954516 DOI: 10.1016/j.bbrc.2020.01.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 01/07/2020] [Indexed: 12/18/2022]
Abstract
Mechanical stimulation is well known to be important for maintaining tissue and organ homeostasis. Here, we found that hydrostatic pressure induced nuclear translocation of a forkhead box O (FOXO) transcription factor DAF-16, in C. elegans within minutes, whereas the removal of this pressure resulted in immediate export of DAF-16 to the cytoplasm. We also monitored DAF-16-dependent transcriptional changes by exposure to 1 MPa pressure for 5 min, and found significant changes in collagen and other genes in a DAF-16 dependent manner. Lifespan was markedly prolonged with exposure to cyclic pressure treatment (1 MPa once a day for 5 min from L1 larvae until death). Furthermore, age-dependent decline in locomotor activity was suppressed by the treatment. In contrast, the nuclear translocation of the yes-associated protein YAP-1 was not induced under the same pressure conditions. Thus, moderate hydrostatic pressure improves ageing progression through activation of DAF-16/FOXO in C. elegans.
Collapse
Affiliation(s)
- Naoshi Watanabe
- Graduate School of Life Sciences, Tohoku University, Sendai, 980-8577, Japan; Faculty of Education, Miyagi University of Education, Sendai, 980-0845, Japan.
| | - Masatoshi Morimatsu
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan.
| | - Ayano Fujita
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Mika Teranishi
- Graduate School of Life Sciences, Tohoku University, Sendai, 980-8577, Japan
| | - Surabhi Sudevan
- Graduate School of Life Sciences, Tohoku University, Sendai, 980-8577, Japan
| | - Masaru Watanabe
- Graduate School of Environmental Studies, Tohoku University, Sendai, 980-8579, Japan; Research Center of Supercritical Fluid Technology, Graduate School of Engineering, Tohoku University, Sendai, 980-8579, Japan
| | - Hiroaki Iwasa
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8519, Japan
| | - Yutaka Hata
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8519, Japan
| | - Hiroyuki Kagi
- Geochemical Research Center, Graduate School of Science, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Masayoshi Nishiyama
- Faculty of Science and Engineering, Kindai University, Osaka, 577-8502, Japan
| | - Keiji Naruse
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan.
| | - Atsushi Higashitani
- Graduate School of Life Sciences, Tohoku University, Sendai, 980-8577, Japan.
| |
Collapse
|
53
|
Yang KJ, Kim JH, Chang YK, Park CW, Kim SY, Hong YA. Inhibition of xanthine oxidoreductase protects against contrast-induced renal tubular injury by activating adenosine monophosphate-activated protein kinase. Free Radic Biol Med 2019; 145:209-220. [PMID: 31560952 DOI: 10.1016/j.freeradbiomed.2019.09.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 09/22/2019] [Indexed: 01/10/2023]
Abstract
Reactive oxygen species (ROS) play a pivotal role in the development of contrast-induced nephropathy (CIN). The inhibition of xanthine oxidoreductase is known to reduce levels of ROS. We investigated whether febuxostat could attenuate oxidative stress via the activation of adenosine monophosphate-activated protein kinase (AMPK) against CIN. In a mouse model of CIN, renal impairment and tubular injury substantially increased, whereas febuxostat attenuated renal injury. Plasma and kidney xanthine oxidoreductase levels were decreased by febuxostat. Febuxostat administration was accompanied by the upregulation of AMPK phosphorylation and the inhibition of nicotinamide-adenine dinucleotide phosphate oxidase (Nox)1 and Nox2, followed by the inhibition of hypoxia-inducible factor-1α (HIF-1α) and heme oxygenase-1 expressions and the suppression of transcription factor forkhead box O (FoxO)1 and FoxO3a phosphorylation. Cell survival was significantly reduced after iohexol administration and febuxostat ameliorated iohexol-induced cell death in proximal tubular (HK-2) cells. Furthermore, febuxostat enhanced AMPK phosphorylation and inhibited Nox1, Nox2, and HIF-1α expression in iohexol-exposed HK-2 cells. Finally, these processes decrease ROS in both in vivo and in vitro models of CIN. AMPK inhibition using small interfering RNA blunted the antioxidative effects of febuxostat in iohexol-treated HK-2 cells. Febuxostat attenuated CIN by modulating oxidative stress through AMPK-NADPH oxidase-HIF-1α signaling.
Collapse
Affiliation(s)
- Keum-Jin Yang
- Clinical Research Institute, Daejeon St. Mary's Hospital, Daejeon, Republic of Korea
| | - Jeong Ho Kim
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoon Kyung Chang
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Cheol Whee Park
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Suk Young Kim
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yu Ah Hong
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
54
|
Hawkins LJ, Hadj-Moussa H, Nguyen VC, Pamenter ME, Storey KB. Naked mole rats activate neuroprotective proteins during hypoxia. JOURNAL OF EXPERIMENTAL ZOOLOGY PART 2019; 331:571-576. [PMID: 31545006 DOI: 10.1002/jez.2321] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/03/2019] [Accepted: 09/05/2019] [Indexed: 12/20/2022]
Abstract
Naked mole rats are a long-lived animal model that age much like humans, but that can also withstand oxidative damage, cancer, neurodegenerative diseases, and severe hypoxic conditions, which is of particular interest to this study. The conditions of their underground burrows result in competition for oxygen consumption, yet despite this oxygen deprivation they emerge unscathed. To understand the mechanisms in place to facilitate neuronal preservation during hypoxia, we investigated the protein levels of well-known cell-stress factors. We found that under hypoxic conditions, nearly half of the proteins measured increased expression in brain, while only a few decreased. Under hypoxic conditions there appeared to be a HIF1α-centered response, where HIF1α and its interactors carbonic anhydrase 9, CITED2, p21/CIP1, and NFκB1, among others, were upregulated. Concurrently, a hypoxia-induced decrease of cytochrome c was consistent with decreased mitochondrial function and protection from apoptosis. The picture that emerges is one of neuroprotection, cell-cycle arrest, and the promotion of antiapoptotic functions, all of which are consistent with conserving energy and maintaining neural integrity under low oxygen levels. These results suggest how this species may be poised to face hypoxia and contribute to its remarkable ability to deal with myriad of other damaging factors and sets the stage for future work on the neuroprotective facilitators we identified.
Collapse
Affiliation(s)
- Liam J Hawkins
- Department of Biology, Carleton University, Ottawa, ON, Canada
| | | | - Vu C Nguyen
- Biology Department, University of Ottawa, Marie-Curie Pvt, Ottawa, ON, Canada
| | - Matthew E Pamenter
- Biology Department, University of Ottawa, Marie-Curie Pvt, Ottawa, ON, Canada.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | | |
Collapse
|
55
|
Fasano C, Disciglio V, Bertora S, Lepore Signorile M, Simone C. FOXO3a from the Nucleus to the Mitochondria: A Round Trip in Cellular Stress Response. Cells 2019; 8:cells8091110. [PMID: 31546924 PMCID: PMC6769815 DOI: 10.3390/cells8091110] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 12/25/2022] Open
Abstract
Cellular stress response is a universal mechanism that ensures the survival or negative selection of cells in challenging conditions. The transcription factor Forkhead box protein O3 (FOXO3a) is a core regulator of cellular homeostasis, stress response, and longevity since it can modulate a variety of stress responses upon nutrient shortage, oxidative stress, hypoxia, heat shock, and DNA damage. FOXO3a activity is regulated by post-translational modifications that drive its shuttling between different cellular compartments, thereby determining its inactivation (cytoplasm) or activation (nucleus and mitochondria). Depending on the stress stimulus and subcellular context, activated FOXO3a can induce specific sets of nuclear genes, including cell cycle inhibitors, pro-apoptotic genes, reactive oxygen species (ROS) scavengers, autophagy effectors, gluconeogenic enzymes, and others. On the other hand, upon glucose restriction, 5′-AMP-activated protein kinase (AMPK) and mitogen activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) -dependent FOXO3a mitochondrial translocation allows the transcription of oxidative phosphorylation (OXPHOS) genes, restoring cellular ATP levels, while in cancer cells, mitochondrial FOXO3a mediates survival upon genotoxic stress induced by chemotherapy. Interestingly, these target genes and their related pathways are diverse and sometimes antagonistic, suggesting that FOXO3a is an adaptable player in the dynamic homeostasis of normal and stressed cells. In this review, we describe the multiple roles of FOXO3a in cellular stress response, with a focus on both its nuclear and mitochondrial functions.
Collapse
Affiliation(s)
- Candida Fasano
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Bari), Italy.
| | - Vittoria Disciglio
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Bari), Italy.
| | - Stefania Bertora
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Bari), Italy.
| | - Martina Lepore Signorile
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Bari), Italy.
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Roma, Italy.
| | - Cristiano Simone
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Bari), Italy.
- Division of Medical Genetics, Department of Biomedical Sciences and Human Oncology (DIMO), University of Bari Aldo Moro, 70124 Bari, Italy.
| |
Collapse
|
56
|
Babosova O, Kapralova K, Raskova Kafkova L, Korinek V, Divoky V, Prchal JT, Lanikova L. Iron chelation and 2-oxoglutarate-dependent dioxygenase inhibition suppress mantle cell lymphoma's cyclin D1. J Cell Mol Med 2019; 23:7785-7795. [PMID: 31517438 PMCID: PMC6815829 DOI: 10.1111/jcmm.14655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/09/2019] [Accepted: 08/10/2019] [Indexed: 01/21/2023] Open
Abstract
The patients with mantle cell lymphoma (MCL) have translocation t(11;14) associated with cyclin D1 overexpression. We observed that iron (an essential cofactor of dioxygenases including prolyl hydroxylases [PHDs]) depletion by deferoxamine blocked MCL cells' proliferation, increased expression of DNA damage marker γH2AX, induced cell cycle arrest and decreased cyclin D1 level. Treatment of MCL cell lines with dimethyloxalylglycine, which blocks dioxygenases involving PHDs by competing with their substrate 2-oxoglutarate, leads to their decreased proliferation and the decrease of cyclin D1 level. We then postulated that loss of EGLN2/PHD1 in MCL cells may lead to down-regulation of cyclin D1 by blocking the degradation of FOXO3A, a cyclin D1 suppressor. However, the CRISPR/Cas9-based loss-of-function of EGLN2/PHD1 did not affect cyclin D1 expression and the loss of FOXO3A did not restore cyclin D1 levels after iron chelation. These data suggest that expression of cyclin D1 in MCL is not controlled by ENGL2/PHD1-FOXO3A pathway and that chelation- and 2-oxoglutarate competition-mediated down-regulation of cyclin D1 in MCL cells is driven by yet unknown mechanism involving iron- and 2-oxoglutarate-dependent dioxygenases other than PHD1. These data support further exploration of the use of iron chelation and 2-oxoglutarate-dependent dioxygenase inhibitors as a novel therapy of MCL.
Collapse
Affiliation(s)
- Olga Babosova
- Department of Cell and Developmental Biology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Katarina Kapralova
- Department of Biology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic.,Division of Hematology & Hematologic Malignancies, Department of Internal Medicine, University of Utah School of Medicine and VAH, Salt Lake City, Utah
| | - Leona Raskova Kafkova
- Department of Biology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Vladimir Korinek
- Department of Cell and Developmental Biology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Vladimir Divoky
- Department of Biology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Josef T Prchal
- Division of Hematology & Hematologic Malignancies, Department of Internal Medicine, University of Utah School of Medicine and VAH, Salt Lake City, Utah
| | - Lucie Lanikova
- Department of Cell and Developmental Biology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic.,Department of Biology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic.,Division of Hematology & Hematologic Malignancies, Department of Internal Medicine, University of Utah School of Medicine and VAH, Salt Lake City, Utah
| |
Collapse
|
57
|
Han J, Shao J, Chen Q, Sun H, Guan L, Li Y, Liu J, Liu H. Transcriptional changes in the hypothalamus, pituitary, and mammary gland underlying decreased lactation performance in mice under heat stress. FASEB J 2019; 33:12588-12601. [PMID: 31480864 DOI: 10.1096/fj.201901045r] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Because of climate change, heat stress (HS) causes more and more impacts on dairy animals to decrease lactation performance. The neuroendocrine system is key in regulating systemic physiological processes and milk synthesis. However, the hypothalamic-pituitary axis response to HS is still unclear. In this study, a group of lactating mice underwent a daily 2-h heat treatment (36°C) for 14 d to explore possible cross-talk between the hypothalamic-pituitary axis and mammary gland under HS. Transcriptome analyses by multitissue RNA-Seq indicated the possible mechanisms of reduced lactation performance in animals under HS. In the hypothalamus, the cAMP signaling pathway was activated to resist neuronal death, and the expression of downstream genes was increased to promote cell survival under HS. Reduced food intake might be caused by down-regulated appetite-related peptide, whereas up-regulated neuropeptide Y acted to attenuate reduced food intake. In pituitary, energy stress from lower food intake might result in reduced secretion of prolactin and growth hormone. Under HS, the mammary gland may undergo hypoxic stress, causing mammary epithelial cell apoptosis. Together, these data showed systemic changes in tissues to accommodate the effects of HS on lactation.-Han, J., Shao, J., Chen, Q., Sun, H., Guan, L., Li, Y., Liu, J., Liu, H. Transcriptional changes in the hypothalamus, pituitary, and mammary gland underlying decreased lactation performance in mice under heat stress.
Collapse
Affiliation(s)
- Jialiang Han
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and
| | - Juanjuan Shao
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and
| | - Qiong Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and
| | - Huizeng Sun
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and.,Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Leluo Guan
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and.,Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Yongxin Li
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and
| | - Jianxin Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and
| | - Hongyun Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China; and
| |
Collapse
|
58
|
Henriques V, Machado S, Link W, Ferreira BI. Monitoring the Transcriptional Activity of FOXO Transcription Factors by Analyzing their Target Genes. Methods Mol Biol 2019; 1890:103-113. [PMID: 30414148 DOI: 10.1007/978-1-4939-8900-3_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
FOXO proteins represent a subfamily of transcription factors that belong to the forkhead family. The study of FOXO target genes can be performed using Real-Time PCR (RT-PCR). The RT-PCR is a sensitive method that allows the detection and quantification of minute amounts of nucleic acids. In RT-PCR the accumulation of the amplicon is detected and measured as the reaction progresses. Here, we describe the application of RT-PCR technique to monitor the transcriptional activity of FOXO transcription factors.
Collapse
Affiliation(s)
- Vanessa Henriques
- Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal
| | - Susana Machado
- Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal
| | - Wolfgang Link
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - Bibiana I Ferreira
- Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal. .,Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal. .,Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal.
| |
Collapse
|
59
|
Ross CI, Shute RJ, Ruby BC, Slivka DR. Skeletal Muscle mRNA Response to Hypobaric and Normobaric Hypoxia After Normoxic Endurance Exercise. High Alt Med Biol 2019; 20:141-149. [DOI: 10.1089/ham.2018.0147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Caleb I. Ross
- Exercise Physiology Lab, University of Nebraska at Omaha, Omaha, Nebraska
| | - Robert J. Shute
- Exercise Physiology Lab, University of Nebraska at Omaha, Omaha, Nebraska
| | - Brent C. Ruby
- Montana Center for Work Physiology and Exercise Metabolism, University of Montana, Missoula, Montana
| | - Dustin R. Slivka
- Exercise Physiology Lab, University of Nebraska at Omaha, Omaha, Nebraska
| |
Collapse
|
60
|
Lee HJ, Feng JH, Sim SM, Lim SS, Lee JY, Suh HW. Effects of resveratrol and oxyresveratrol on hippocampal cell death induced by kainic acid. Anim Cells Syst (Seoul) 2019; 23:246-252. [PMID: 31489245 PMCID: PMC6711029 DOI: 10.1080/19768354.2019.1620853] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/25/2019] [Accepted: 05/15/2019] [Indexed: 12/26/2022] Open
Abstract
In the present study, we have examined the possible neuroprotective effects of resveratrol and oxyresveratrol against kainic-acid (KA)-induced hippocampal neuronal cell death. Either resveratrol or oxyresvertrol was orally administered 30 min prior to intracerebroventricular (i.c.v.) administration with KA (0.05 μg). Oral pretreatment with oxyresveratrol (50 mg/kg) significantly protected KA-induced hippocampal CA3 neuronal cell death. However, the same dose (50 mg/kg) or a higher dose (100 mg/kg) pretreatment with resveratrol did not affect KA-induced hippocampal neuronal cell death. Furthermore, the i.c.v. pretreatment with 30 μg of oxyresveratrol or resveratrol did not show the protective effect against KA-induced hippocampal neuronal cell death. In the immunohistochemical analysis, FoxO3a and pFoxO3a expressions in the hippocampal CA3 region were significantly increased 30 min after KA administration. Oral pretreatment with oxyresveratrol (50 mg/kg) significantly reduced KA-induced Forkhead homeobox type O3a (FoxO3a) and pFoxO3a expression in CA3 region of the hippocampus, suggesting that oxyresveratrol may exert a neuroprotective effect against KA-induced hippocampal neuronal cell death by reducing the levels of FoxO3a and pFoxO3a protein expression in the hippocampal CA3 region. Furthermore, it is suggested that the neuroprotective effect of orally administered oxyresveratrol against KA-induced neurotoxicity might be possibly mediated by some metabolites rather than direct action of oxyresveratrol on the central nervous system.
Collapse
Affiliation(s)
- Hee-Jung Lee
- Department of Pharmacology, Institute of Natural Medicine, College of Medicine Hallym University, Chuncheon, Republic of Korea
| | - Jing-Hui Feng
- Department of Pharmacology, Institute of Natural Medicine, College of Medicine Hallym University, Chuncheon, Republic of Korea
| | - Su-Min Sim
- Department of Pharmacology, Institute of Natural Medicine, College of Medicine Hallym University, Chuncheon, Republic of Korea
| | - Soon-Sung Lim
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea
| | - Jae-Yong Lee
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Hong-Won Suh
- Department of Pharmacology, Institute of Natural Medicine, College of Medicine Hallym University, Chuncheon, Republic of Korea
| |
Collapse
|
61
|
Hypoxia induces the dormant state in oocytes through expression of Foxo3. Proc Natl Acad Sci U S A 2019; 116:12321-12326. [PMID: 31147464 DOI: 10.1073/pnas.1817223116] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In mammals, most immature oocytes remain dormant in the primordial follicles to ensure the longevity of female reproductive life. A precise understanding of mechanisms underlying the dormancy is important for reproductive biology and medicine. In this study, by comparing mouse oogenesis in vivo and in vitro, the latter of which bypasses the primordial follicle stage, we defined the gene-expression profile representing the dormant state of oocytes. Overexpression of constitutively active FOXO3 partially reproduced the dormant state in vitro. Based on further gene-expression analysis, we found that a hypoxic condition efficiently induced the dormant state in vitro. The effect of hypoxia was severely diminished by disruption of the Foxo3 gene and inhibition of hypoxia-inducible factors. Our findings provide insights into the importance of environmental conditions and their effectors for establishing the dormant state.
Collapse
|
62
|
Transcriptional regulators CITED2 and PU.1 cooperate in maintaining hematopoietic stem cells. Exp Hematol 2019; 73:38-49.e7. [PMID: 30986495 DOI: 10.1016/j.exphem.2019.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/27/2019] [Accepted: 03/13/2019] [Indexed: 12/27/2022]
Abstract
Reduced expression of the transcription factor PU.1 is frequently associated with development of acute myeloid leukemia (AML), whereas elevated levels of CITED2 (CBP/p300-interacting-transactivator-with-an-ED-rich-tail 2) enhance maintenance of both normal and leukemic hematopoietic stem and progenitor cells (HSPCs). Recent findings indicate that PU.1 and CITED2 act in the same gene regulatory network. We therefore examined a potential synergistic effect of simultaneous PU.1 downregulation and CITED2 upregulation on stem cell biology and AML pathogenesis. We found that simultaneous PU.1/CITED2 deregulation in human CD34+ cord blood (CB) cells, as well as CITED2 upregulation in preleukemic murine PU.1-knockdown (PU.1KD/KD) bone marrow cells, significantly increased the maintenance of HSPCs compared with the respective deregulation of either factor alone. Increased replating capacity of PU.1KD/KD/CITED2 cells in in vitro assays eventually resulted in outgrowth of transformed cells, while upregulation of CITED2 in PU.1KD/KD cells enhanced their engraftment in in vivo transplantation studies without affecting leukemic transformation. Transcriptional analysis of CD34+ CB cells with combined PU.1/CITED2 alterations revealed a set of differentially expressed genes that highly correlated with gene signatures found in various AML subtypes. These findings illustrate that combined PU.1/CITED2 deregulation induces a transcriptional program that promotes HSPC maintenance, which might be a prerequisite for malignant transformation.
Collapse
|
63
|
Li L, Kang H, Zhang Q, D'Agati VD, Al-Awqati Q, Lin F. FoxO3 activation in hypoxic tubules prevents chronic kidney disease. J Clin Invest 2019; 129:2374-2389. [PMID: 30912765 DOI: 10.1172/jci122256] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Acute kidney injury (AKI) can lead to chronic kidney disease (CKD) if injury is severe and/or repair is incomplete. However, the pathogenesis of CKD following renal ischemic injury is not fully understood. Capillary rarefaction and tubular hypoxia are common findings during the AKI to CKD transition. We investigated the tubular stress response to hypoxia and demonstrated that a stress responsive transcription factor, FoxO3, was regulated by prolyl hydroxylase. Hypoxia inhibited FoxO3 prolyl hydroxylation and FoxO3 degradation, thus leading to FoxO3 accumulation and activation in tubular cells. Hypoxia-activated Hif-1α contributed to FoxO3 activation and functioned to protect kidneys, as tubular deletion of Hif-1α decreased hypoxia-induced FoxO3 activation, and resulted in more severe tubular injury and interstitial fibrosis following ischemic injury. Strikingly, tubular deletion of FoxO3 during the AKI to CKD transition aggravated renal structural and functional damage leading to a more profound CKD phenotype. We showed that tubular deletion of FoxO3 resulted in decreased autophagic response and increased oxidative injury, which may explain renal protection by FoxO3. Our study indicates that in the hypoxic kidney, stress responsive transcription factors can be activated for adaptions to counteract hypoxic insults, thus attenuating CKD development.
Collapse
Affiliation(s)
- Ling Li
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Huimin Kang
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA.,Department of Pediatrics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Qing Zhang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Qais Al-Awqati
- Department of Internal Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Fangming Lin
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
64
|
4-O-Methylascochlorin inhibits the prolyl hydroxylation of hypoxia-inducible factor-1α, which is attenuated by ascorbate. J Antibiot (Tokyo) 2019; 72:271-281. [PMID: 30796332 DOI: 10.1038/s41429-019-0157-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 01/14/2019] [Accepted: 01/31/2019] [Indexed: 12/27/2022]
Abstract
4-O-Methylascochlorin (MAC), a methylated derivative of ascochlorin, was previously shown to promote the accumulation of hypoxia-inducible factor (HIF)-1α in human breast adenocarcinoma MCF-7 cells. In the present study, we further investigated the effects of MAC on the expression and function of HIF-1α in human fibrosarcoma HT-1080 cells. MAC promoted the accumulation of the HIF-1α protein without affecting its constitutive mRNA expression and augmented the transcriptional activation of HIF target genes. Ascorbate, but not N-acetylcysteine, attenuated MAC-mediated HIF-1α accumulation. MAC-induced increases in HIF-1α transcriptional activity were also attenuated by ascorbate. MAC inhibited the hydroxylation of HIF-1α at the proline 564 residue, while it was reversed by ascorbate. MAC slightly decreased the intracellular concentration of ascorbate. The present results demonstrated that MAC promoted the accumulation of HIF-1α by preventing prolyl hydroxylation, and ascorbate attenuated the MAC-mediated inhibition of HIF-1α prolyl hydroxylation.
Collapse
|
65
|
Tian T, Li X, Zhang J. mTOR Signaling in Cancer and mTOR Inhibitors in Solid Tumor Targeting Therapy. Int J Mol Sci 2019; 20:ijms20030755. [PMID: 30754640 PMCID: PMC6387042 DOI: 10.3390/ijms20030755] [Citation(s) in RCA: 403] [Impact Index Per Article: 67.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 01/28/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022] Open
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) pathway plays a crucial role in regulation of cell survival, metabolism, growth and protein synthesis in response to upstream signals in both normal physiological and pathological conditions, especially in cancer. Aberrant mTOR signaling resulting from genetic alterations from different levels of the signal cascade is commonly observed in various types of cancers. Upon hyperactivation, mTOR signaling promotes cell proliferation and metabolism that contribute to tumor initiation and progression. In addition, mTOR also negatively regulates autophagy via different ways. We discuss mTOR signaling and its key upstream and downstream factors, the specific genetic changes in the mTOR pathway and the inhibitors of mTOR applied as therapeutic strategies in eight solid tumors. Although monotherapy and combination therapy with mTOR inhibitors have been extensively applied in preclinical and clinical trials in various cancer types, innovative therapies with better efficacy and less drug resistance are still in great need, and new biomarkers and deep sequencing technologies will facilitate these mTOR targeting drugs benefit the cancer patients in personalized therapy.
Collapse
Affiliation(s)
- Tian Tian
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| | - Xiaoyi Li
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| | - Jinhua Zhang
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| |
Collapse
|
66
|
Liu F, Huang X, Luo Z, He J, Haider F, Song C, Peng L, Chen T, Wu B. Hypoxia-Activated PI3K/Akt Inhibits Oxidative Stress via the Regulation of Reactive Oxygen Species in Human Dental Pulp Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6595189. [PMID: 30728888 PMCID: PMC6343138 DOI: 10.1155/2019/6595189] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/19/2018] [Accepted: 10/30/2018] [Indexed: 01/02/2023]
Abstract
In order to use stem cells as a resource for tissue regeneration, it is necessary to induce expansion in vitro. However, during culture, stem cells often lose functional properties and become senescent. Increasing evidence indicates that hypoxic preconditioning with physiological oxygen concentration can maintain the functional properties of stem cells in vitro. The purpose of the current study was to test the hypothesis that hypoxic preconditioning with physiological oxygen concentration can maintain the functional properties of stem cells in culture by reducing oxidative stress. In vitro studies were performed in primary human dental pulp cells (hDPCs). Reduced levels of oxidative stress and increased cellular "stemness" in response to physiological hypoxia were dependent upon the expression of reactive oxygen species (ROS). Subsequently, RNA-sequencing analysis revealed the increased expression of phosphoinositide 3-kinase (PI3K)/Akt signaling in culture, a pathway which regulates oxidative stress. Furthermore, we found evidence that PI3K/Akt signaling might affect intracellular ROS production by negatively regulating expression of the downstream protein Forkhead Box Protein O1 (FOXO1) and Caspase 3. Collectively, our data show that the PI3K/Akt pathway is activated in response to hypoxia and inhibits oxidative stress in a ROS-dependent manner. This study identified redox-mediated hypoxic preconditioning regulatory mechanisms that may be significant for tissue regeneration.
Collapse
Affiliation(s)
- Fei Liu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- College of Stomatology, Southern Medical University, Guangzhou 510515, China
- International Medical Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Xin Huang
- Department of Stomatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Zhenhua Luo
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | - Jingjun He
- International Medical Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Farhan Haider
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ci Song
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ling Peng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ting Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Buling Wu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
67
|
Identification of Novel Therapeutic Targets for Pulmonary Arterial Hypertension. Int J Mol Sci 2018; 19:ijms19124081. [PMID: 30562953 PMCID: PMC6321293 DOI: 10.3390/ijms19124081] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 12/13/2018] [Accepted: 12/13/2018] [Indexed: 12/29/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) and chronic thromboembolic pulmonary hypertension (CTEPH) are fatal diseases; however, their pathogenesis still remains to be elucidated. We have recently screened novel pathogenic molecules and have performed drug discovery targeting those molecules. Pulmonary artery smooth muscle cells (PASMCs) in patients with PAH (PAH-PASMCs) have high proliferative properties like cancer cells, which leads to thickening and narrowing of distal pulmonary arteries. Thus, we conducted a comprehensive analysis of PAH-PASMCs and lung tissues to search for novel pathogenic proteins. We validated the pathogenic role of the selected proteins by using tissue-specific knockout mice. To confirm its clinical significance, we used patient-derived blood samples to evaluate the potential as a biomarker for diagnosis and prognosis. Finally, we conducted a high throughput screening and found inhibitors for the pathogenic proteins.
Collapse
|
68
|
Curcumin pretreatment protects against hypoxia/reoxgenation injury via improvement of mitochondrial function, destabilization of HIF-1α and activation of Epac1-Akt pathway in rat bone marrow mesenchymal stem cells. Biomed Pharmacother 2018; 109:1268-1275. [PMID: 30551377 DOI: 10.1016/j.biopha.2018.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/01/2018] [Accepted: 11/01/2018] [Indexed: 11/22/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) possess promising therapeutic effects and have been considered as a highly desirable agent for tissue injury treatment. However, little survived cells after transplanting due to severe relocated conditions (characterized by prolonged hypoxia and oxidative stress) lead to hampered benefits of BMSCs-based cell therapy. Curcumin, a natural dietary product, has attracted increasing attention owing to its profound pharmacologic properties. Here, we report the protective effects of curcumin pretreatment in BMSCs against hypoxia and reoxygenation (H/R) triggered injury, which mimick ischemia/reperfusion in vivo. We found that curcumin pretreatment remarkably inhibited H/R-induced cell viability loss, cell nuclei condensation, LDH leakage, as well as caspase-3 activity increase in BMSCs. Furthermore, curcumin pretreatment prevented H/R-induced mitochondrial dysfunction through expediting adenosine triphosphate production and suppressing reactive oxygen species accumulation and mitochondrial membrane potential decline. In addition, curcumin pretreatment notably induced HIF-1α destabilization, Epac1 and Akt activation, and Erk1/2 and p38 deactivation. However, Epac1 inhibitor ESI-09 obviously restrained the increase of p-Akt induced by curcumin, but not p-Erk1/2 or p-p38, and abrogated the protective effect of curcumin on BMSCs' survival and arrested cell cycle in G0/G1 phase. Taken together, these results demonstrated that curcumin pretreatment conferred BMSCs the ability to survive from H/R injury, which might attribute to its protection on mitochondrial function, destabilization of HIF-1α and activation of Epac1-Akt signaling pathway. Thus, this study provides more pharmacologic aspects of curcumin, and suggests that pre-conditioning of BMSCs with curcumin could serve as an attractive approach for facilitating cell therapy in tissue repair treatment.
Collapse
|
69
|
Chen F, Wang RJ, Li GZ, Zhang Y, Yu S, Liu YF, Chen XY, Hou SK. miRNA array analysis of plasma miRNA alterations in rats exposed to a high altitude hypoxic environment. Mol Med Rep 2018; 18:5502-5510. [PMID: 30365091 PMCID: PMC6236226 DOI: 10.3892/mmr.2018.9570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 03/20/2017] [Indexed: 11/06/2022] Open
Abstract
In the present study, the microRNA (miRNA) expression profiles of rats exposed to high altitude hypoxia and normal conditions were obtained from miRNA array analysis. Bioinformatics analyses, including the use of the Gene Oncology and Kyoto Encyclopedia of Genes and Genomes databases, were used to identify the genes and pathways, which were specifically associated with high altitude hypoxic environment‑associated miRNAs. A total of 26 miRNAs were differentially expressed in the two groups, comprising six upregulated and 20 downregulated miRNAs. In the present study, a novel pattern of upregulated miRNAs and their associated pathways were constructed, including proteoglycans in cancer, spliceosome, gluamatergic synapse, glycolysis/gluconeogenesis, Foxo, cGMP‑PKG and p53 signaling pathways, which may provide novel targets for diagnosing and understanding the mechanism of high altitude hypoxia‑induced disease.
Collapse
Affiliation(s)
- Feng Chen
- Graduate School, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Ren-Jie Wang
- Department of Neurosurgery, Logistics University of Chinese People's Armed Police Force, Tianjin 300162, P.R. China
| | - Guang-Zong Li
- Key Laboratory of Disaster and Emergency Rescue Medicine in People's Liberation Army, Logistics University of Chinese People's Armed Police Force, Tianjin 300162, P.R. China
| | - Yi Zhang
- Key Laboratory of Disaster and Emergency Rescue Medicine in People's Liberation Army, Logistics University of Chinese People's Armed Police Force, Tianjin 300162, P.R. China
| | - Shuo Yu
- Key Laboratory of Disaster and Emergency Rescue Medicine in People's Liberation Army, Logistics University of Chinese People's Armed Police Force, Tianjin 300162, P.R. China
| | - Ying-Fu Liu
- Department of Cell Biology, Logistics University of Chinese People's Armed Police Forces, Tianjin 300309, P.R. China
| | - Xiao-Yi Chen
- Department of Cell Biology, Logistics University of Chinese People's Armed Police Forces, Tianjin 300309, P.R. China
| | - Shi-Ke Hou
- Graduate School, Tianjin Medical University, Tianjin 300070, P.R. China
| |
Collapse
|
70
|
Sabzevary-Ghahfarokhi M, Shirzad H, Rafieian-Kopaei M, Ghatreh-Samani M, Shohan M. The Role of Inflammatory Cytokines in Creating T Cell Exhaustion in Cancer. Cancer Biother Radiopharm 2018; 33:267-273. [DOI: 10.1089/cbr.2018.2449] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Milad Sabzevary-Ghahfarokhi
- Department of Microbiology and Immunology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hedayatollah Shirzad
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mahmoud Rafieian-Kopaei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mahdi Ghatreh-Samani
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mojtaba Shohan
- Department of Microbiology and Immunology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
71
|
Kikuchi N, Satoh K, Kurosawa R, Yaoita N, Elias-Al-Mamun M, Siddique MAH, Omura J, Satoh T, Nogi M, Sunamura S, Miyata S, Saito Y, Hoshikawa Y, Okada Y, Shimokawa H. Selenoprotein P Promotes the Development of Pulmonary Arterial Hypertension: Possible Novel Therapeutic Target. Circulation 2018; 138:600-623. [PMID: 29636330 DOI: 10.1161/circulationaha.117.033113] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/28/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND Excessive proliferation and apoptosis resistance of pulmonary artery smooth muscle cells (PASMCs) are key mechanisms of pulmonary arterial hypertension (PAH). Despite the multiple combination therapy, a considerable number of patients develop severe pulmonary hypertension (PH) because of the lack of diagnostic biomarker and antiproliferative therapies for PASMCs. METHODS Microarray analyses were used to identify a novel therapeutic target for PAH. In vitro experiments, including lung and serum samples from patients with PAH, cultured PAH-PASMCs, and high-throughput screening of 3336 low-molecular-weight compounds, were used for mechanistic study and exploring a novel therapeutic agent. Five genetically modified mouse strains, including PASMC-specific selenoprotein P (SeP) knockout mice and PH model rats, were used to study the role of SeP and therapeutic capacity of the compounds for the development of PH in vivo. RESULTS Microarray analysis revealed a 32-fold increase in SeP in PAH-PASMCs compared with control PASMCs. SeP is a widely expressed extracellular protein maintaining cellular metabolism. Immunoreactivity of SeP was enhanced in the thickened media of pulmonary arteries in PAH. Serum SeP levels were also elevated in patients with PH compared with controls, and high serum SeP predicted poor outcome. SeP-knockout mice ( SeP-/-) exposed to chronic hypoxia showed significantly reduced right ventricular systolic pressure, right ventricular hypertrophy, and pulmonary artery remodeling compared with controls. In contrast, systemic SeP-overexpressing mice showed exacerbation of hypoxia-induced PH. Furthermore, PASMC-specific SeP-/- mice showed reduced hypoxia-induced PH compared with controls, whereas neither liver-specific SeP knockout nor liver-specific SeP-overexpressing mice showed significant differences with controls. Altogether, protein levels of SeP in the lungs were associated with the development of PH. Mechanistic experiments demonstrated that SeP promotes PASMC proliferation and resistance to apoptosis through increased oxidative stress and mitochondrial dysfunction, which were associated with activated hypoxia-inducible factor-1α and dysregulated glutathione metabolism. It is important to note that the high-throughput screening of 3336 compounds identified that sanguinarine, a plant alkaloid with antiproliferative effects, reduced SeP expression and proliferation in PASMCs and ameliorated PH in mice and rats. CONCLUSIONS These results indicate that SeP promotes the development of PH, suggesting that it is a novel biomarker and therapeutic target of the disorder.
Collapse
MESH Headings
- Animals
- Antihypertensive Agents/pharmacology
- Apoptosis
- Arterial Pressure/drug effects
- Benzophenanthridines/pharmacology
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Humans
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/prevention & control
- Hypoxia/complications
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Isoquinolines/pharmacology
- Male
- Mice, Knockout
- Mitochondria, Muscle/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Oxidative Stress
- Pulmonary Artery/metabolism
- Pulmonary Artery/physiopathology
- Rats, Sprague-Dawley
- Selenoprotein P/metabolism
- Signal Transduction
- Vascular Remodeling/drug effects
Collapse
Affiliation(s)
- Nobuhiro Kikuchi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
- Research Fellow of Japan Society for the Promotion of Science, Tokyo (N.K., R.K.)
| | - Kimio Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Ryo Kurosawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
- Research Fellow of Japan Society for the Promotion of Science, Tokyo (N.K., R.K.)
| | - Nobuhiro Yaoita
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Md Elias-Al-Mamun
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Mohammad Abdul Hai Siddique
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Junichi Omura
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Taijyu Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Masamichi Nogi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Shinichiro Sunamura
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Satoshi Miyata
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| | - Yoshiro Saito
- Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan (Y.S.)
| | - Yasushi Hoshikawa
- Department of Thoracic Surgery, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan (Y.H., Y.O.)
| | - Yoshinori Okada
- Department of Thoracic Surgery, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan (Y.H., Y.O.)
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (N.K., K.S., R.K., S.M., N.Y., M.E.-A.-M., M.A.H.S., J.O., T.S., M.N., S.S., H.S.)
| |
Collapse
|
72
|
Tang Q, Wu H, Lei J, Yi C, Xu W, Lan W, Yang F, Liu C. HIF1α deletion facilitates adipose stem cells to repair renal fibrosis in diabetic mice. In Vitro Cell Dev Biol Anim 2018; 54:272-286. [PMID: 29511913 DOI: 10.1007/s11626-018-0231-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/08/2018] [Indexed: 12/21/2022]
Abstract
Adipose stem cell (ASC) transplantation is a promising therapeutic strategy for diabetic renal fibrosis. Hypoxia-inducible factor 1α (HIF1α) is a negative regulatory factor of mitochondrial function. In the current study, we aimed to explore if HIF1α deletion protects against hyperglycemia-induced ASC damage and enhances the therapeutic efficiency of ASCs in diabetic renal fibrosis. Our data indicated that HIF1α was upregulated in ASCs in response to high glucose stimulation. Higher HIF1α expression was associated with ASC apoptosis and proliferation arrest. Loss of HIF1α activated mitophagy protecting ASCs against high glucose-induced apoptosis via preserving mitochondrial function. Transplanting HIF1α-deleted ASCs in db/db mice improved the abnormalities in glucose metabolic parameters, including the levels of glucose, insulin, C-peptide, HbA1c, and inflammatory markers. In addition, the engraftment of HIF1α-modified ASCs also reversed renal function, decreased renal hypertrophy, and ameliorated renal histological changes in db/db mice. Functional studies confirmed that HIF1α-modified ASCs reduced renal fibrosis. Collectively, our results demonstrate that ASCs may be a promising therapeutic treatment for ameliorating diabetes and the development of renal fibrosis and that the loss of HIF1α in ASCs may further increase the efficiency of stem cell-based therapy. These findings provide a new understanding about the protective effects of HIF1α silencing on ASCs and offer a new strategy for promoting the therapeutic efficacy of ASCs in diabetic renal fibrosis.
Collapse
Affiliation(s)
- Qun Tang
- Department of Pathology, Medical school, Hunan University of Chinese Medicine, No.300, Xueshi Road, Hanpu kejiao Park, Yuelu District, Changsha, Hunan Province, 410208, China
| | - Hua Wu
- Hunan Furong Judicial Authentication Center, The Second People's Hospital of Hunan Province, Changsha, 410007, China
| | - Jiushi Lei
- Department of Pathology, Medical school, Hunan University of Chinese Medicine, No.300, Xueshi Road, Hanpu kejiao Park, Yuelu District, Changsha, Hunan Province, 410208, China
| | - Chun Yi
- Department of Pathology, Medical school, Hunan University of Chinese Medicine, No.300, Xueshi Road, Hanpu kejiao Park, Yuelu District, Changsha, Hunan Province, 410208, China
| | - Wenfeng Xu
- Department of Pathology, Medical school, Hunan University of Chinese Medicine, No.300, Xueshi Road, Hanpu kejiao Park, Yuelu District, Changsha, Hunan Province, 410208, China
| | - Wenqu Lan
- Department of Pathology, Medical school, Hunan University of Chinese Medicine, No.300, Xueshi Road, Hanpu kejiao Park, Yuelu District, Changsha, Hunan Province, 410208, China
| | - Fang Yang
- Department of Pathology, Medical school, Hunan University of Chinese Medicine, No.300, Xueshi Road, Hanpu kejiao Park, Yuelu District, Changsha, Hunan Province, 410208, China
| | - Chunyan Liu
- Department of Pathology, Medical school, Hunan University of Chinese Medicine, No.300, Xueshi Road, Hanpu kejiao Park, Yuelu District, Changsha, Hunan Province, 410208, China.
| |
Collapse
|
73
|
Xie Y, Jiang D, Xiao J, Fu C, Zhang Z, Ye Z, Zhang X. Ischemic preconditioning attenuates ischemia/reperfusion-induced kidney injury by activating autophagy via the SGK1 signaling pathway. Cell Death Dis 2018; 9:338. [PMID: 29497029 PMCID: PMC5832808 DOI: 10.1038/s41419-018-0358-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 12/12/2022]
Abstract
Ischemic preconditioning (IPC) has a strong renoprotective effect during renal ischemia/reperfusion (I/R) injury that is thought to relate to autophagy. However, the role of autophagy during IPC-afforded renoprotection and the precise mechanisms involved are unknown. In this study, an in vitro hypoxia/reoxygenation (H/R) model was established in which oxygen and glucose deprivation (OGD) was applied to renal cells for 15 h followed by reoxygenation under normal conditions for 30 min, 2 h or 6 h; transient OGD and subsequent reoxygenation were implemented before prolonged H/R injury to achieve hypoxic preconditioning (HPC). 3-Methyladenine (3-MA) was used to inhibit autophagy. In a renal I/R injury model, rats were subjected to 40 min of renal ischemia followed by 6 h, 12 h or 24 h of reperfusion. IPC was produced by four cycles of ischemia (8 min each) followed by 5 min of reperfusion prior to sustained ischemia. We found that IPC increased LC3II and Beclin-1 levels and decreased SQSTM/p62 and cleaved caspase-3 levels in a time-dependent manner during renal I/R injury, as well as increased the number of intracellular double-membrane vesicles in injured renal cells. IPC-induced renal protection was efficiently attenuated by pretreatment with 5 mM 3-MA. Pretreatment with IPC also dynamically affected the expression of SGK1/FOXO3a/HIF-1α signaling components. Moreover, knocking down SGK1 expression significantly downregulated phosphorylated-FOXO3a (p-FOXO3a)/FOXO3 and HIF-1α, suppressed LC3II and Beclin-1 levels, increased SQSTM/p62 and cleaved caspase-3 levels, and abolished the protective effect of IPC against I/R-induced renal damage. SGK1 overexpression efficiently increased p-FOXO3a/FOXO3 and HIF-1α levels, promoted the autophagy flux and enhanced the protective effect mediated by HPC. Furthermore, FOXO3a overexpression decreased HIF-1α protein levels, inhibited HIF-1α transcriptional activity and reduced the protective effect of IPC. Our study indicates that IPC can ameliorate renal I/R injury by promoting autophagy through the SGK1 pathway.
Collapse
Affiliation(s)
- Ying Xie
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
| | - Daofang Jiang
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
| | - Jing Xiao
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, China
| | - Chensheng Fu
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, China
| | - Zhenxing Zhang
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, China
| | - Zhibin Ye
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
| | - Xiaoli Zhang
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, China. .,Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China.
| |
Collapse
|
74
|
Ahn H, Kim H, Abdul R, Kim Y, Sim J, Choi D, Paik SS, Shin SJ, Kim DH, Jang K. Overexpression of Forkhead Box O3a and Its Association With Aggressive Phenotypes and Poor Prognosis in Human Hepatocellular Carcinoma. Am J Clin Pathol 2018; 149:117-127. [PMID: 29365018 DOI: 10.1093/ajcp/aqx132] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVES Recent research has demonstrated that forkhead box O3a (FoxO3a) may function as an oncogenic transcription factor. We sought to validate the clinicopathologic significance of FoxO3a expression in hepatocellular carcinoma (HCC). METHODS Western blotting and immunohistochemistry were used to determine FoxO3a expression. In vitro cell proliferation and migration assays were performed in a HepG2 cell line. RESULTS FoxO3a was overexpressed in 121 (64.71%) cases of HCC. FoxO3a overexpression was associated with aggressive phenotypes of HCC, such as histologic grade (P < .001), stage (P = .031), and small vessel invasion (P < .001). FoxO3a overexpression was also correlated with poor disease-free survival in both univariate and multivariate survival analyses (P = .001 and P = .018, respectively). Downregulation of FoxO3a in a HepG2 cell line inhibited cell proliferation and migration. CONCLUSIONS These results suggest a role for FoxO3a in HCC progression and support the potential use as a prognostic biomarker.
Collapse
Affiliation(s)
- Hyein Ahn
- Department of Pathology, Konyang University College of Medicine, Daejeon, Korea
| | - Hyunsung Kim
- Departments of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Rehman Abdul
- Departments of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Yesul Kim
- Departments of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Jongmin Sim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dongho Choi
- Departments of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Seung Sam Paik
- Departments of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Su-Jin Shin
- Departments of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Dong-Hoon Kim
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kiseok Jang
- Departments of Pathology, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
75
|
mTOR Cross-Talk in Cancer and Potential for Combination Therapy. Cancers (Basel) 2018; 10:cancers10010023. [PMID: 29351204 PMCID: PMC5789373 DOI: 10.3390/cancers10010023] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 12/20/2022] Open
Abstract
The mammalian Target of Rapamycin (mTOR) pathway plays an essential role in sensing and integrating a variety of exogenous cues to regulate cellular growth and metabolism, in both physiological and pathological conditions. mTOR functions through two functionally and structurally distinct multi-component complexes, mTORC1 and mTORC2, which interact with each other and with several elements of other signaling pathways. In the past few years, many new insights into mTOR function and regulation have been gained and extensive genetic and pharmacological studies in mice have enhanced our understanding of how mTOR dysfunction contributes to several diseases, including cancer. Single-agent mTOR targeting, mostly using rapalogs, has so far met limited clinical success; however, due to the extensive cross-talk between mTOR and other pathways, combined approaches are the most promising avenues to improve clinical efficacy of available therapeutics and overcome drug resistance. This review provides a brief and up-to-date narrative on the regulation of mTOR function, the relative contributions of mTORC1 and mTORC2 complexes to cancer development and progression, and prospects for mTOR inhibition as a therapeutic strategy.
Collapse
|
76
|
van Doeselaar S, Burgering BMT. FOXOs Maintaining the Equilibrium for Better or for Worse. Curr Top Dev Biol 2018; 127:49-103. [PMID: 29433740 DOI: 10.1016/bs.ctdb.2017.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A paradigm shift is emerging within the FOXO field and accumulating evidence indicates that we need to reappreciate the role of FOXOs, at least in cancer development. Here, we discuss the possibility that FOXOs are both tumor suppressors as well as promoters of tumor progression. This is mostly dependent on the biological context. Critical to this dichotomous role is the notion that FOXOs are central in preserving cellular homeostasis in redox control, genomic stability, and protein turnover. From this perspective, a paradoxical role in both suppressing and enhancing tumor progression can be reconciled. As many small molecules targeting the PI3K pathway are developed by big pharmaceutical companies and/or are in clinical trial, we will discuss what the consequences may be for the context-dependent role of FOXOs in tumor development in treatment options based on active PI3K signaling in tumors.
Collapse
Affiliation(s)
- Sabina van Doeselaar
- Molecular Cancer Research, Center Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Boudewijn M T Burgering
- Molecular Cancer Research, Center Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
77
|
Yadav RK, Chauhan AS, Zhuang L, Gan B. FoxO transcription factors in cancer metabolism. Semin Cancer Biol 2018; 50:65-76. [PMID: 29309929 DOI: 10.1016/j.semcancer.2018.01.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 12/28/2017] [Accepted: 01/04/2018] [Indexed: 12/21/2022]
Abstract
FoxO transcription factors serve as the central regulator of cellular homeostasis and are tumor suppressors in human cancers. Recent studies have revealed that, besides their classic functions in promoting cell death and inducing cell cycle arrest, FoxOs also regulate cancer metabolism, an emerging hallmark of cancer. In this review, we summarize the regulatory mechanisms employed to control FoxO activities in the context of cancer biology, and discuss FoxO function in metabolism reprogramming in cancer and interaction with other key cancer metabolism pathways. A deeper understanding of FoxOs in cancer metabolism may reveal novel therapeutic opportunities in cancer treatment.
Collapse
Affiliation(s)
- Raj Kumar Yadav
- Department of Experimental Radiation Oncology, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Anoop Singh Chauhan
- Department of Experimental Radiation Oncology, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA.
| | - Li Zhuang
- Department of Experimental Radiation Oncology, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; Department of Molecular and Cellular Oncology, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, 1515 Holcombe Blvd, Houston, TX 77030, USA.
| |
Collapse
|
78
|
Bayen S, Saini S, Gaur P, Duraisamy AJ, Kumar Sharma A, Pal K, Vats P, Singh SB. PRMT1 promotes hyperglycemia in a FoxO1-dependent manner, affecting glucose metabolism, during hypobaric hypoxia exposure, in rat model. Endocrine 2018; 59:151-163. [PMID: 29128891 DOI: 10.1007/s12020-017-1463-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 08/22/2017] [Indexed: 01/02/2023]
Abstract
PURPOSE High-altitude (HA) environment causes changes in cellular metabolism among unacclimatized humans. Previous studies have revealed that insulin-dependent activation of protein kinase B (Akt) regulates metabolic processes via discrete transcriptional effectors. Moreover, protein arginine methyltransferase (PRMT)1-dependent arginine modification of forkhead box other (FoxO)1 protein interferes with Akt-dependent phosphorylation. The present study was undertaken to test the involvement of PRMT1 on FoxO1 activation during hypobaric hypoxia (HH) exposure in rat model. METHODS Samples were obtained from normoxia control (NC) and HH-exposed (H) rats, subdivided according to the duration of HH exposure. To explore the specific role played by PRMT1 during HH exposure, samples from 1d pair-fed (PF) NC, 1d acute hypoxia-exposed (AH) placebo-treated, and 1d AH TC-E-5003-treated rats were investigated. Quantitative reverse transcriptase polymerase chain reaction (RT-qPCR) was performed to determine expressions of glycolytic, gluconeogenic enzymes, and insulin response regulating genes. Immuno-blot and enzyme linked immunosorbent assay (ELISA) were used for insulin response regulating proteins. Nuclear translocation of FoxO1 was analyzed using deoxyribonucleic acid (DNA)-binding ELISA kit. RESULTS We observed HH-induced increase in glycolytic enzyme expressions in hepatic tissue unlike hypothalamic tissue. PRMT1 expression increased during HH exposure, causing insulin resistance and resulting increase in FoxO1 nuclear translocation, leading to hyperglycemia. Conversely, PRMT1 inhibitor treatment promoted inhibition of FoxO1 activity and increase in glucose uptake during HH exposure leading to reduction in blood-glucose and hepatic glycogen levels. CONCLUSIONS PRMT1 might have a potential importance as a therapeutic target for the treatment of HH-induced maladies.
Collapse
Affiliation(s)
- Susovon Bayen
- Endocrinology & Metabolism Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Supriya Saini
- Endocrinology & Metabolism Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Priya Gaur
- Endocrinology & Metabolism Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Arul Joseph Duraisamy
- Endocrinology & Metabolism Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Alpesh Kumar Sharma
- Endocrinology & Metabolism Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Karan Pal
- Endocrinology & Metabolism Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Praveen Vats
- Endocrinology & Metabolism Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India.
| | - Shashi Bala Singh
- Department of Applied Physiology, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India
| |
Collapse
|
79
|
Alvarez-Garcia O, Matsuzaki T, Olmer M, Masuda K, Lotz MK. Age-related reduction in the expression of FOXO transcription factors and correlations with intervertebral disc degeneration. J Orthop Res 2017; 35:2682-2691. [PMID: 28430387 PMCID: PMC5650945 DOI: 10.1002/jor.23583] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/07/2017] [Indexed: 02/04/2023]
Abstract
Aging is a main risk factor for intervertebral disc (IVD) degeneration, the main cause of low back pain. FOXO transcription factors are important regulators of tissue homeostasis and longevity. Here, we determined the expression pattern of FOXO in healthy and degenerated human IVD and the associations with IVD degeneration during mouse aging. FOXO expression was assessed by immunohistochemistry in normal and degenerated human IVD samples and in cervical and lumbar IVD from 6-, 12-, 24-, and 36-month-old C57BL/6J mice. Mouse spines were graded for key histological features of disc degeneration in all the time points and expression of two key FOXO downstream targets, sestrin 3 (SESN3) and superoxide dismutase (SOD2), was assessed by immunohistochemistry. Histological analysis revealed that FOXO proteins are expressed in all compartments of human and mouse IVD. Expression of FOXO1 and FOXO3, but not FOXO4, was significantly deceased in human degenerated discs. In mice, degenerative changes in the lumbar spine were seen at 24 and 36 months of age whereas cervical IVD showed increased histopathological scores at 36 months. FOXO expression was significantly reduced in lumbar IVD at 12-, 24-, and 36-month-old mice and in cervical IVD at 36-month-old mice when compared with the 6-month-old group. The reduction of FOXO expression in lumbar IVD was concomitant with a decrease in the expression of SESN3 and SOD2. These findings suggest that reduced FOXO expression occurs in lumbar IVD during aging and precedes the major histopathological changes associated with lumbar IVD degeneration. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2682-2691, 2017.
Collapse
Affiliation(s)
- Oscar Alvarez-Garcia
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Tokio Matsuzaki
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Merissa Olmer
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Koichi Masuda
- Department of Orthopedic Surgery, University of California-San Diego, San Diego, CA, USA
| | - Martin K. Lotz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
80
|
Möllerherm H, Meier K, Schmies K, Fuhrmann H, Naim HY, von Köckritz-Blickwede M, Branitzki-Heinemann K. Differentiation and Functionality of Bone Marrow-Derived Mast Cells Depend on Varying Physiologic Oxygen Conditions. Front Immunol 2017; 8:1665. [PMID: 29250065 PMCID: PMC5714875 DOI: 10.3389/fimmu.2017.01665] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/14/2017] [Indexed: 01/17/2023] Open
Abstract
Mast cells (MCs) are long-living multifunctional innate immune cells that originate from hematopoietic precursors and specifically differentiate in the destination tissue, e.g., skin, respiratory mucosa, intestine, where they mediate immune cell recruitment and antimicrobial defense. In vivo these tissues have characteristic physiological oxygen levels that are considerably lower than the atmospheric oxygen conditions (159 mmHg, 21% O2; 5% CO2) traditionally used to differentiate MCs and to study their functionality in vitro. Only little is known about the impact of physiological oxygen conditions on the differentiation process of MCs. This study aimed to characterize the differentiation of immature murine bone marrow-derived MCs under physioxia in vitro (7% O2; 53 mmHg; 5% CO2). Bone marrow-derived suspension cells were differentiated in the presence of interleukin-3 with continuous, non-invasive determination of the oxygen level using a Fibox4-PSt3 measurement system without technique-caused oxygen consumption. Trypan blue staining confirmed cellular viability during the specified period. Interestingly, MCs cultivated at 7% O2 showed a significantly delayed differentiation rate defined by CD117-positive cells, analyzed by flow cytometry, and reached >95% CD117 positive population at day 32 after isolation. Importantly, MCs differentiated under physioxia displayed a decreased transcript expression level of hif-1α and selected target genes vegf, il-6, and tnf-α, but an increase of foxo3 and vhl expression compared to MCs cultivated under normoxia. Moreover, the production of reactive oxygen species as well as the amount of intracellular stored histamine was significantly lower in MCs differentiated under low oxygen levels, which might have consequences for their function such as immunomodulation of other immune cells. These results show for the first time that physioxia substantially affect maturation and the properties of MCs and highlight the need to study their function under physiologically relevant oxygen conditions.
Collapse
Affiliation(s)
- Helene Möllerherm
- Department of Physiological Chemistry, University for Veterinary Medicine Hannover, Hanover, Germany
| | - Karsten Meier
- Department of Physiological Chemistry, University for Veterinary Medicine Hannover, Hanover, Germany
| | - Kathrin Schmies
- Department of Physiological Chemistry, University for Veterinary Medicine Hannover, Hanover, Germany
| | - Herbert Fuhrmann
- Faculty of Veterinary Medicine, Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | - Hassan Y Naim
- Department of Physiological Chemistry, University for Veterinary Medicine Hannover, Hanover, Germany
| | - Maren von Köckritz-Blickwede
- Department of Physiological Chemistry, University for Veterinary Medicine Hannover, Hanover, Germany.,Research Center for Emerging Infections and Zoonoses (RIZ), University for Veterinary Medicine Hannover, Hanover, Germany
| | - Katja Branitzki-Heinemann
- Department of Physiological Chemistry, University for Veterinary Medicine Hannover, Hanover, Germany
| |
Collapse
|
81
|
Hou T, Li Z, Zhao Y, Zhu WG. Mechanisms controlling the anti-neoplastic functions of FoxO proteins. Semin Cancer Biol 2017; 50:101-114. [PMID: 29155239 DOI: 10.1016/j.semcancer.2017.11.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 10/18/2017] [Accepted: 11/14/2017] [Indexed: 02/06/2023]
Abstract
The Forkhead box O (FoxO) proteins comprise a family of evolutionarily conserved transcription factors that predominantly function as tumor suppressors. These proteins assume diverse roles in the cellular anti-neoplastic response, including regulation of apoptosis and autophagy, cancer metabolism, cell-cycle arrest, oxidative stress and the DNA damage response. More recently, FoxO proteins have been implicated in cancer immunity and cancer stem-cell (CSC) homeostasis. Interestingly, in some sporadic sub-populations, FoxO protein function may also be manipulated by factors such as β-catenin whereby they instead can facilitate cancer progression via maintenance of CSC properties or promoting drug resistance or metastasis and invasion. This review highlights the essential biological functions of FoxOs and explores the areas that may be exploited in FoxO protein signaling pathways in the development of novel cancer therapeutic agents.
Collapse
Affiliation(s)
- Tianyun Hou
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Zhiming Li
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Ying Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wei-Guo Zhu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
82
|
Chen YF, Pandey S, Day CH, Chen YF, Jiang AZ, Ho TJ, Chen RJ, Padma VV, Kuo WW, Huang CY. Synergistic effect of HIF-1α and FoxO3a trigger cardiomyocyte apoptosis under hyperglycemic ischemia condition. J Cell Physiol 2017; 233:3660-3671. [PMID: 29030976 DOI: 10.1002/jcp.26235] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 09/29/2017] [Indexed: 01/19/2023]
Abstract
Cardiomyocyte death is an important pathogenic feature of ischemia and heart failure. Through this study, we showed the synergistic role of HIF-1α and FoxO3a in cardiomyocyte apoptosis subjected to hypoxia plus elevated glucose levels. Using gene specific small interfering RNAs (siRNA), semi-quantitative reverse transcriptase polymerase chain reaction (RT-PCR), Western blot, immunofluorescence, nuclear and cytosolic localization and TUNEL assay techniques, we determined that combined function of HIF-1α and FoxO3a under high glucose plus hypoxia condition lead to enhanced expression of BNIP3 inducing cardiomyocyte death. Our results highlighted the importance of the synergistic role of HIF-1α and FoxO3a in cardiomyocyte death which may add insight into therapeutic approaches to pathophysiology associated with ischemic diabetic cardiomyopathies.
Collapse
Affiliation(s)
- Ya-Fang Chen
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Obstetrics and Gynecology, Taichung Veteran's General Hospital, Taichung, Taiwan
| | - Sudhir Pandey
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | | | - Yu-Feng Chen
- Section of Cardiology, Yuan Rung Hospital, Yuanlin, Taiwan
| | - Ai-Zhi Jiang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Tsung-Jung Ho
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Ray-Jade Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Vijaya V Padma
- Department of Biotechnology, Bharathiar University, Coimbatore, India
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.,Faculty of Applied Sciences, Ton Duc Thang University, Tan Phong Ward, Ho Chi Minh City, Vietnam.,Department of Biological Science and Technology, Asia University, Taichung, Taiwan
| |
Collapse
|
83
|
Abstract
The present review aimed to assess the networks of transcription factors regulating the Oct4 expression in mice. Through a comprehensive analysis of the binding sites and the interrelationships of the transcription factors of Oct4, it is found that transcription factors of Oct4 form three regulating complexes centered by Oct4-Sox2, Nanog, and Lrh1. They bind on CR4, CR2, and CR1 regions of Oct4 promoter/enhancer, respectively, to activate Oct4 transcription synergistically. This article also discusses the mechanisms of fine-tuning the Oct4 expression. These findings have important implications in the field of stem cell and developmental biology.
Collapse
Affiliation(s)
- Yu-Qiang Li
- Marine College, Shandong University (Weihai) , Weihai, China
| |
Collapse
|
84
|
Li L, Zviti R, Ha C, Wang ZV, Hill JA, Lin F. Forkhead box O3 (FoxO3) regulates kidney tubular autophagy following urinary tract obstruction. J Biol Chem 2017; 292:13774-13783. [PMID: 28705935 DOI: 10.1074/jbc.m117.791483] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/11/2017] [Indexed: 12/20/2022] Open
Abstract
Autophagy has been shown to be important for normal homeostasis and adaptation to stress in the kidney. Yet, the molecular mechanisms regulating renal epithelial autophagy are not fully understood. Here, we explored the role of the stress-responsive transcription factor forkhead box O3 (FoxO3) in mediating injury-induced proximal tubular autophagy in mice with unilateral ureteral obstruction (UUO). We show that following UUO, FoxO3 is activated and displays nuclear expression in the hypoxic proximal tubules exhibiting high levels of autophagy. Activation of FoxO3 by mutating phosphorylation sites to enhance its nuclear expression induces profound autophagy in cultured renal epithelial cells. Conversely, deleting FoxO3 in mice results in fewer numbers of autophagic cells in the proximal tubules and reduced ratio of the autophagy-related protein LC3-II/I in the kidney post-UUO. Interestingly, autophagic cells deficient in FoxO3 contain lower numbers of autophagic vesicles per cell. Analyses of individual cells treated with autophagic inhibitors to sequentially block the autophagic flux suggest that FoxO3 stimulates the formation of autophagosomes to increase autophagic capacity but has no significant effect on autophagosome-lysosome fusion or autolysosomal clearance. Furthermore, in kidneys with persistent UUO for 7 days, FoxO3 activation increases the abundance of mRNA and protein levels of the core autophagy-related (Atg) proteins including Ulk1, Beclin-1, Atg9A, Atg4B, and Bnip3, suggesting that FoxO3 may function to maintain components of the autophagic machinery that would otherwise be consumed during prolonged autophagy. Taken together, our findings indicate that FoxO3 activation can both induce and maintain autophagic activities in renal epithelial cells in response to injury from urinary tract obstruction.
Collapse
Affiliation(s)
- Ling Li
- From the Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York 10032 and
| | - Ronald Zviti
- From the Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York 10032 and
| | - Catherine Ha
- From the Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York 10032 and
| | - Zhao V Wang
- the Departments of Internal Medicine (Cardiology) and.,Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Joseph A Hill
- the Departments of Internal Medicine (Cardiology) and.,Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Fangming Lin
- From the Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York 10032 and
| |
Collapse
|
85
|
Link W, Fernandez-Marcos PJ. FOXO transcription factors at the interface of metabolism and cancer. Int J Cancer 2017. [PMID: 28631330 DOI: 10.1002/ijc.30840] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diabetes refers to a group of metabolic diseases characterized by impaired insulin signalling and high blood glucose. A growing body of epidemiological evidence links diabetes to several types of cancer but the underlying molecular mechanisms are poorly understood. The signalling cascade connecting insulin and FOXO proteins provides a compelling example for a conserved pathway at the interface between insulin signalling and cancer. FOXOs are transcription factors that orchestrate programs of gene expression known to control a variety of processes in response to cellular stress. Genes regulated by this family of proteins are involved in the regulation of cellular energy production, oxidative stress resistance and cell viability and proliferation. Accordingly, FOXO factors have been shown to play an important role in the suppression of tumour growth and in the regulation of metabolic homeostasis. There is emerging evidence that deregulation of FOXO factors might account for the association between insulin resistance-related metabolic disorders and cancer.
Collapse
Affiliation(s)
- Wolfgang Link
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, room 2.22, Faro, 8005-139, Portugal.,Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Campus de Gambelas, Faro, 8005-139, Portugal.,Algarve Biomedical Center (ABC), University of Algarve, Campus de Gambelas, Faro, 8005-139, Portugal
| | | |
Collapse
|
86
|
Lee HJ, Jung YH, Choi GE, Ko SH, Lee SJ, Lee SH, Han HJ. BNIP3 induction by hypoxia stimulates FASN-dependent free fatty acid production enhancing therapeutic potential of umbilical cord blood-derived human mesenchymal stem cells. Redox Biol 2017; 13:426-443. [PMID: 28704726 PMCID: PMC5508529 DOI: 10.1016/j.redox.2017.07.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/29/2017] [Accepted: 07/03/2017] [Indexed: 02/08/2023] Open
Abstract
Mitophagy under hypoxia is an important factor for maintaining and regulating stem cell functions. We previously demonstrated that fatty acid synthase (FASN) induced by hypoxia is a critical lipid metabolic factor determining the therapeutic efficacy of umbilical cord blood-derived human mesenchymal stem cells (UCB-hMSCs). Therefore, we investigated the mechanism of a major mitophagy regulator controlling lipid metabolism and therapeutic potential of UCB-hMSCs. This study revealed that Bcl2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3)-dependent mitophagy is important for reducing mitochondrial reactive oxygen species accumulation, anti-apoptosis, and migration under hypoxia. And, BNIP3 expression was regulated by CREB binding protein-mediated transcriptional actions of HIF-1α and FOXO3. Silencing of BNIP3 suppressed free fatty acid (FFA) synthesis regulated by SREBP1/FASN pathway, which is involved in UCB-hMSC apoptosis via caspases cleavage and migration via cofilin-1-mediated F-actin reorganization in hypoxia. Moreover, reduced mouse skin wound-healing capacity of UCB-hMSC with hypoxia pretreatment by BNIP3 silencing was recovered by palmitic acid. Collectively, our findings suggest that BNIP3-mediated mitophagy under hypoxia leads to FASN-induced FFA synthesis, which is critical for therapeutic potential of UCB-hMSCs with hypoxia pretreatment. BNIP3 induction by hypoxia mainly controls mitophagy and mitochondrial ROS production in UCB-hMSCs. BNIP3 silencing impairs UCB-hMSC functions such as survival, migration and free fatty acid production under hypoxia. BNIP3 silencing suppresses SREBP1/FASN-mediated free fatty acid production via ROS regulation under hypoxia. BNIP3 silencing decreased skin wound healing potential of hypoxia-pretreated UCB-hMSCs. Palmitic acid addition recovers decreased therapeutic potential of UCB-hMSCs by BNIP3 silencing.
Collapse
Affiliation(s)
- Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Republic of Korea
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Republic of Korea
| | - Gee Euhn Choi
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Republic of Korea
| | - So Hee Ko
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Republic of Korea
| | - Sei-Jung Lee
- Department of Pharmaceutical Engineering, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea; Departments of Biochemistry, Soonchunhyang University College of Medicine, Cheonan 330-930, Republic of Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
87
|
Kim LC, Cook RS, Chen J. mTORC1 and mTORC2 in cancer and the tumor microenvironment. Oncogene 2017; 36:2191-2201. [PMID: 27748764 PMCID: PMC5393956 DOI: 10.1038/onc.2016.363] [Citation(s) in RCA: 319] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/04/2016] [Accepted: 08/15/2016] [Indexed: 02/06/2023]
Abstract
The mammalian target of rapamycin (mTOR) is a crucial signaling node that integrates environmental cues to regulate cell survival, proliferation and metabolism, and is often deregulated in human cancer. mTOR kinase acts in two functionally distinct complexes, mTOR complex 1 (mTORC1) and 2 (mTORC2), whose activities and substrate specificities are regulated by complex co-factors. Deregulation of this centralized signaling pathway has been associated with a variety of human diseases including diabetes, neurodegeneration and cancer. Although mTORC1 signaling has been extensively studied in cancer, recent discoveries indicate a subset of human cancers harboring amplifications in mTORC2-specific genes as the only actionable genomic alterations, suggesting a distinct role for mTORC2 in cancer as well. This review will summarize recent advances in dissecting the relative contributions of mTORC1 versus mTORC2 in cancer, their role in tumor-associated blood vessels and tumor immunity, and provide an update on mTOR inhibitors.
Collapse
Affiliation(s)
- Laura C. Kim
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232
| | - Rebecca S. Cook
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232
| | - Jin Chen
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232
- Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, TN 37212
| |
Collapse
|
88
|
Chen Z, Soutto M, Rahman B, Fazili MW, Peng D, Blanca Piazuelo M, Chen H, Kay Washington M, Shyr Y, El-Rifai W. Integrated expression analysis identifies transcription networks in mouse and human gastric neoplasia. Genes Chromosomes Cancer 2017; 56:535-547. [PMID: 28281307 DOI: 10.1002/gcc.22456] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 02/24/2017] [Accepted: 02/27/2017] [Indexed: 12/16/2022] Open
Abstract
Gastric cancer (GC) is a leading cause of cancer-related deaths worldwide. The Tff1 knockout (KO) mouse model develops gastric lesions that include low-grade dysplasia (LGD), high-grade dysplasia (HGD), and adenocarcinomas. In this study, we used Affymetrix microarrays gene expression platforms for analysis of molecular signatures in the mouse stomach [Tff1-KO (LGD) and Tff1 wild-type (normal)] and human gastric cancer tissues and their adjacent normal tissue samples. Combined integrated bioinformatics analysis of mouse and human datasets indicated that 172 genes were consistently deregulated in both human gastric cancer samples and Tff1-KO LGD lesions (P < .05). Using Ingenuity pathway analysis, these genes mapped to important transcription networks that include MYC, STAT3, β-catenin, RELA, NFATC2, HIF1A, and ETS1 in both human and mouse. Further analysis demonstrated activation of FOXM1 and inhibition of TP53 transcription networks in human gastric cancers but not in Tff1-KO LGD lesions. Using real-time RT-PCR, we validated the deregulated expression of several genes (VCAM1, BGN, CLDN2, COL1A1, COL1A2, COL3A1, EpCAM, IFITM1, MMP9, MMP12, MMP14, PDGFRB, PLAU, and TIMP1) that map to altered transcription networks in both mouse and human gastric neoplasia. Our study demonstrates significant similarities in deregulated transcription networks in human gastric cancer and gastric tumorigenesis in the Tff1-KO mouse model. The data also suggest that activation of MYC, STAT3, RELA, and β-catenin transcription networks could be an early molecular step in gastric carcinogenesis.
Collapse
Affiliation(s)
- Zheng Chen
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, 37232.,Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Mohammed Soutto
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, 37232.,Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Bushra Rahman
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Muhammad W Fazili
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - DunFa Peng
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Maria Blanca Piazuelo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Gastroenterology, Hepatology, & Nutrition, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Heidi Chen
- Center of Quantitative Sciences, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN, 37232
| | - M Kay Washington
- Department of Pathology, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN, 37232
| | - Yu Shyr
- Center of Quantitative Sciences, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN, 37232
| | - Wael El-Rifai
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, 37232.,Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232.,Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, 37232
| |
Collapse
|
89
|
Kietzmann T. Metabolic zonation of the liver: The oxygen gradient revisited. Redox Biol 2017; 11:622-630. [PMID: 28126520 PMCID: PMC5257182 DOI: 10.1016/j.redox.2017.01.012] [Citation(s) in RCA: 345] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 02/06/2023] Open
Abstract
The liver has a multitude of functions which are necessary to maintain whole body homeostasis. This requires that various metabolic pathways can run in parallel in the most efficient manner and that futile cycles are kept to a minimum. To a large extent this is achieved due to a functional specialization of the liver parenchyma known as metabolic zonation which is often lost in liver diseases. Although this phenomenon is known for about 40 years, the underlying regulatory pathways are not yet fully elucidated. The physiologically occurring oxygen gradient was considered to be crucial for the appearance of zonation; however, a number of reports during the last decade indicating that β-catenin signaling, and the hedgehog (Hh) pathway contribute to metabolic zonation may have shifted this view. In the current review we connect these new observations with the concept that the oxygen gradient within the liver acinus is a regulator of zonation. This is underlined by a number of facts showing that the β-catenin and the Hh pathway can be modulated by the hypoxia signaling system and the hypoxia-inducible transcription factors (HIFs). Altogether, we provide a view by which the dynamic interplay between all these pathways can drive liver zonation and thus contribute to its physiological function.
Collapse
Affiliation(s)
- Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland.
| |
Collapse
|
90
|
Acetylation promotes TyrRS nuclear translocation to prevent oxidative damage. Proc Natl Acad Sci U S A 2017; 114:687-692. [PMID: 28069943 DOI: 10.1073/pnas.1608488114] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Tyrosyl-tRNA synthetase (TyrRS) is well known for its essential aminoacylation function in protein synthesis. Recently, TyrRS has been shown to translocate to the nucleus and protect against DNA damage due to oxidative stress. However, the mechanism of TyrRS nuclear localization has not yet been determined. Herein, we report that TyrRS becomes highly acetylated in response to oxidative stress, which promotes nuclear translocation. Moreover, p300/CBP-associated factor (PCAF), an acetyltransferase, and sirtuin 1 (SIRT1), a NAD+-dependent deacetylase, regulate the nuclear localization of TyrRS in an acetylation-dependent manner. Oxidative stress increases the level of PCAF and decreases the level of SIRT1 and deacetylase activity, all of which promote the nuclear translocation of hyperacetylated TyrRS. Furthermore, TyrRS is primarily acetylated on the K244 residue near the nuclear localization signal (NLS), and acetylation inhibits the aminoacylation activity of TyrRS. Molecular dynamics simulations have shown that the in silico acetylation of K244 induces conformational changes in TyrRS near the NLS, which may promote the nuclear translocation of acetylated TyrRS. Herein, we show that the acetylated K244 residue of TyrRS protects against DNA damage in mammalian cells and zebrafish by activating DNA repair genes downstream of transcription factor E2F1. Our study reveals a previously unknown mechanism by which acetylation regulates an aminoacyl-tRNA synthetase, thus affecting the repair pathways for damaged DNA.
Collapse
|
91
|
Choi S, Chung H, Hong H, Kim SY, Kim SE, Seoh JY, Moon CM, Yang EG, Oh ES. Inflammatory hypoxia induces syndecan-2 expression through IL-1β-mediated FOXO3a activation in colonic epithelia. FASEB J 2016; 31:1516-1530. [PMID: 28031321 DOI: 10.1096/fj.201601098r] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/12/2016] [Indexed: 02/06/2023]
Abstract
Chronic inflammation is known to be a key causative factor in tumor progression, but we do not yet fully understand the molecular mechanism through which inflammation leads to cancer. Here, we report that the dextran sulfate sodium (DSS)-induced mouse model of chronic colitis is associated with increases in the serum level of IL-1β and the colonic epithelial expression of the cell-surface heparan sulfate proteoglycan, syndecan-2. We further show that IL-1β stimulated the transcription of syndecan-2 via NF-κB-dependent FOXO3a activation in CCD841CoN normal colonic epithelial cells and early-stage HT29 colon cancer cells. Inflammatory hypoxia was observed in the colonic epithelia of mice with chronic colitis, suggesting that hypoxic stress is involved in the regulation of syndecan-2 expression. Consistently, experimental inflammatory hypoxia induced hypoxia inducible factor-1α-dependent FOXO3a expression and the p38 MAPK-mediated nuclear localization of FOXO3a. FOXO3a directly mediated syndecan-2 expression in both cell lines and the colonic epithelia of mice with DSS-induced colitis. Moreover, syndecan-2 expression was detected in azoxymethane/DSS-induced colon tumors. Together, these data demonstrate that inflammatory hypoxia up-regulates syndecan-2 via the IL-1β-NF-κB-FOXO3a pathway. These findings provide new mechanistic insights into inflammatory hypoxia-mediated syndecan-2 expression to connect chronic inflammation and the development of colon cancer.-Choi, S., Chung, H., Hong, H., Kim, S. Y., Kim, S.-E., Seoh, J.-Y., Moon, C. M., Yang, E. G., Oh, E.-S. Inflammatory hypoxia induces syndecan-2 expression through IL-1β-mediated FOXO3a activation in colonic epithelia.
Collapse
Affiliation(s)
- Sojoong Choi
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Heesung Chung
- Department of Life Sciences, Ewha Womans University, Seoul, South Korea; .,The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Heejeong Hong
- Department of Life Sciences, Ewha Womans University, Seoul, South Korea.,The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - So Yeon Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Seong-Eun Kim
- Department of Internal Medicine, School of Medicine, Ewha Womans University, Seoul, South Korea; and
| | - Ju-Young Seoh
- Department of Microbiology, Graduate School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Chang Mo Moon
- Department of Internal Medicine, School of Medicine, Ewha Womans University, Seoul, South Korea; and
| | - Eun Gyeong Yang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, South Korea;
| | - Eok-Soo Oh
- Department of Life Sciences, Ewha Womans University, Seoul, South Korea; .,The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| |
Collapse
|
92
|
Hagenbuchner J, Rupp M, Salvador C, Meister B, Kiechl-Kohlendorfer U, Müller T, Geiger K, Sergi C, Obexer P, Ausserlechner MJ. Nuclear FOXO3 predicts adverse clinical outcome and promotes tumor angiogenesis in neuroblastoma. Oncotarget 2016; 7:77591-77606. [PMID: 27769056 PMCID: PMC5363607 DOI: 10.18632/oncotarget.12728] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 10/03/2016] [Indexed: 12/30/2022] Open
Abstract
Neuroblastoma is the most frequent, extracranial solid tumor in children with still poor prognosis in stage IV disease. In this study, we analyzed FOXO3-phosphorylation and cellular localization in tumor biopsies and determined the function of this homeostasis regulator in vitro and in vivo. FOXO3-phosphorylation at threonine-32 (T32) and nuclear localization in biopsies significantly correlated with stage IV disease. DNA-damaging drugs induced nuclear accumulation of FOXO3, which was associated with elevated T32-phosphorylation in stage IV-derived neuroblastoma cells, thereby reflecting the in situ results. In contrast, hypoxic conditions repressed PKB-activity and caused dephosphorylation of FOXO3 in both, stroma-like SH-EP and high-stage-derived STA-NB15 cells. The activation of an ectopically-expressed FOXO3 in these cells reduced viability at normoxia, but promoted growth at hypoxic conditions and elevated VEGF-C-expression. In chorioallantoic membrane (CAM) assays STA-NB15 tumors with ectopic FOXO3 showed increased micro-vessel formation and, when xenografted into nude mice, a gene-dosage-dependent effect of FOXO3 in high-stage STA-NB15 cells became evident: low-level activation increased tumor-vascularization, whereas hyper-activation repressed tumor growth.The combined data suggest that, depending on the mode and intensity of activation, cellular FOXO3 acts as a homeostasis regulator promoting tumor growth at hypoxic conditions and tumor angiogenesis in high-stage neuroblastoma.
Collapse
Affiliation(s)
- Judith Hagenbuchner
- Departments of Pediatrics II, Medical University Innsbruck, Innsbruck, Austria
| | - Martina Rupp
- Departments of Pediatrics II, Medical University Innsbruck, Innsbruck, Austria
- Pediatrics I, Medical University Innsbruck, Innsbruck, Austria
| | | | | | | | - Thomas Müller
- Pediatrics I, Medical University Innsbruck, Innsbruck, Austria
| | | | - Consolato Sergi
- Walter C. Mackenzie Centre, University of Alberta, Edmonton, Canada
| | - Petra Obexer
- Departments of Pediatrics II, Medical University Innsbruck, Innsbruck, Austria
- Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Michael J. Ausserlechner
- Pediatrics I, Medical University Innsbruck, Innsbruck, Austria
- Tyrolean Cancer Research Institute, Innsbruck, Austria
| |
Collapse
|
93
|
Brucker DP, Maurer GD, Harter PN, Rieger J, Steinbach JP. FOXO3a orchestrates glioma cell responses to starvation conditions and promotes hypoxia-induced cell death. Int J Oncol 2016; 49:2399-2410. [DOI: 10.3892/ijo.2016.3760] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/21/2016] [Indexed: 11/06/2022] Open
|
94
|
Liu X, Cai X, Zhang D, Xu C, Xiao W. Zebrafish foxo3b Negatively Regulates Antiviral Response through Suppressing the Transactivity of irf3 and irf7. THE JOURNAL OF IMMUNOLOGY 2016; 197:4736-4749. [PMID: 27815423 DOI: 10.4049/jimmunol.1601187] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 10/06/2016] [Indexed: 12/25/2022]
Abstract
Forkhead box O (FOXO)3, a member of the FOXO family of transcription factors, plays key roles in various cellular processes, including development, longevity, reproduction, and metabolism. Recently, FOXO3 has also been shown to be involved in modulating the immune response. However, how FOXO3 regulates immunity and the underlying mechanisms are still largely unknown. In this study, we show that zebrafish (Danio rerio) foxo3b, an ortholog of mammalian FOXO3, is induced by polyinosinic-polycytidylic acid stimulation and spring viremia of carp virus (SVCV) infection. We found that foxo3b interacted with irf3 and irf7 to inhibit ifr3/irf7 transcriptional activity, thus resulting in suppression of SVCV or polyinosinic-polycytidylic acid-induced IFN activation. By suppressing expression of key antiviral genes, foxo3b negatively regulated the cellular antiviral response. Furthermore, upon SVCV infection, the expression of the key antiviral genes was significantly enhanced in foxo3b-null zebrafish larvae compared with wild-type larvae. Additionally, the replication of SVCV was inhibited in foxo3b-null zebrafish larvae, leading to a higher survival rate. Our findings suggest that by suppressing irf3/irf7 activity, zebrafish foxo3b negatively regulates the antiviral response, implicating the vital role of the FOXO gene family in innate immunity.
Collapse
Affiliation(s)
- Xing Liu
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; and
| | - Xiaolian Cai
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; and
| | - Dawei Zhang
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; and
| | - Chenxi Xu
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; and
| | - Wuhan Xiao
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; and .,State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China
| |
Collapse
|
95
|
Webb AE, Kundaje A, Brunet A. Characterization of the direct targets of FOXO transcription factors throughout evolution. Aging Cell 2016; 15:673-85. [PMID: 27061590 PMCID: PMC4933671 DOI: 10.1111/acel.12479] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2016] [Indexed: 12/20/2022] Open
Abstract
FOXO transcription factors (FOXOs) are central regulators of lifespan across species, yet they also have cell‐specific functions, including adult stem cell homeostasis and immune function. Direct targets of FOXOs have been identified genome‐wide in several species and cell types. However, whether FOXO targets are specific to cell types and species or conserved across cell types and throughout evolution remains uncharacterized. Here, we perform a meta‐analysis of direct FOXO targets across tissues and organisms, using data from mammals as well as Caenorhabditis elegans and Drosophila. We show that FOXOs bind cell type‐specific targets, which have functions related to that particular cell. Interestingly, FOXOs also share targets across different tissues in mammals, and the function and even the identity of these shared mammalian targets are conserved in invertebrates. Evolutionarily conserved targets show enrichment for growth factor signaling, metabolism, stress resistance, and proteostasis, suggesting an ancestral, conserved role in the regulation of these processes. We also identify candidate cofactors at conserved FOXO targets that change in expression with age, including CREB and ETS family factors. This meta‐analysis provides insight into the evolution of the FOXO network and highlights downstream genes and cofactors that may be particularly important for FOXO's conserved function in adult homeostasis and longevity.
Collapse
Affiliation(s)
- Ashley E. Webb
- Department of Genetics Stanford University 300 Pasteur Drive Stanford CA 94305 USA
| | - Anshul Kundaje
- Department of Genetics Stanford University 300 Pasteur Drive Stanford CA 94305 USA
| | - Anne Brunet
- Department of Genetics Stanford University 300 Pasteur Drive Stanford CA 94305 USA
- Glenn Laboratories for the Biology of Aging at Stanford Stanford CA 94305 USA
| |
Collapse
|
96
|
Feng CC, Lin CC, Lai YP, Chen TS, Marthandam Asokan S, Lin JY, Lin KH, Viswanadha VP, Kuo WW, Huang CY. Hypoxia suppresses myocardial survival pathway through HIF-1α-IGFBP-3-dependent signaling and enhances cardiomyocyte autophagic and apoptotic effects mainly via FoxO3a-induced BNIP3 expression. Growth Factors 2016; 34:73-86. [PMID: 27366871 DOI: 10.1080/08977194.2016.1191480] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The HIF-1α transcriptional factor and the BH-3 only protein BNIP3 are known to play fundamental roles in response to hypoxia. The objective of this research is to investigate the molecular mechanisms and the correlation of HIF-1α, BNIP3 and IGFBP-3 in hypoxia-induced cardiomyocytes injuries. Heart-derived H9c2 cells and neonatal rat ventricular myocytes (NRVMs) were incubated in normoxic or hypoxic conditions. Hypoxia increased HIF-1α expression and activated the downstream BNIP3 and IGFBP-3 thereby triggered mitochondria-dependent apoptosis. Moreover, IGF1R/PI3K/Akt signaling was attenuated by HIF-1α-dependent IGFBP-3 expression to enhance hypoxia-induced apoptosis. Autophagy suppression with 3-methyladenine or siATG5 or siBeclin-1 significantly decreased myocardial apoptosis under hypoxia. Knockdown of FoxO3a or BNIP3 significantly abrogated hypoxia-induced autophagy and mitochondria-dependent apoptosis. Moreover, prolonged-hypoxia induced HIF-1α stimulated BNIP3 and enhanced IGFBP-3 activation to inhibit IGF1R/PI3K/Akt survival pathway and mediate mitochondria-dependent cardiomyocyte apoptosis. HIF-1α and FoxO3a blockage are sufficient to annul the change of excessive hypoxia of hearts.
Collapse
Affiliation(s)
- Chih-Chung Feng
- a Graduate Institute of Clinical Medical Science, Graduate Institute of Basic Medical Science, China Medical University , Taichung , Taiwan
| | - Chien-Chung Lin
- b Orthopaedic Department, Armed Forces General Hospital , Taichung , Taiwan
| | - Yi-Ping Lai
- c Graduate Institute of Basic Medical Science, China Medical University , Taichung , Taiwan
| | - Tung-Sheng Chen
- c Graduate Institute of Basic Medical Science, China Medical University , Taichung , Taiwan
- d Biomaterials Translational Research Center, China Medical University Hospital , Taichung , Taiwan
| | | | - Jing-Ying Lin
- e Department of Nursing , Central Taiwan University of Science and Technology , Taichung , Taiwan
| | - Kuan-Ho Lin
- f Emergency Department, China Medical University Hospital , Taichung , Taiwan
| | | | - Wei-Wen Kuo
- h Department of Biological Science and Technology , China Medical University , Taichung , Taiwan
| | - Chih-Yang Huang
- c Graduate Institute of Basic Medical Science, China Medical University , Taichung , Taiwan
- i Graduate Institute of Chinese Medical Science, China Medical University , Taichung , Taiwan , and
- j Department of Health and Nutrition Biotechnology , Asia University , Taichung , Taiwan
| |
Collapse
|
97
|
Cummins EP, Keogh CE. Respiratory gases and the regulation of transcription. Exp Physiol 2016; 101:986-1002. [DOI: 10.1113/ep085715] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 05/23/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Eoin P. Cummins
- School of Medicine; University College Dublin; Belfield 4 Dublin Ireland
| | - Ciara E. Keogh
- School of Medicine; University College Dublin; Belfield 4 Dublin Ireland
| |
Collapse
|
98
|
Lang M, Wang X, Wang H, Dong J, Lan C, Hao J, Huang C, Li X, Yu M, Yang Y, Yang S, Ren H. Arsenic trioxide plus PX-478 achieves effective treatment in pancreatic ductal adenocarcinoma. Cancer Lett 2016; 378:87-96. [PMID: 27212442 DOI: 10.1016/j.canlet.2016.05.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 05/12/2016] [Accepted: 05/17/2016] [Indexed: 02/06/2023]
Abstract
Arsenic trioxide (ATO) has been selected as a promising treatment not only in leukemia but also in solid tumors. Previous studies showed that the cytotoxicity of ATO mainly depends on the induction of reactive oxygen species. However, ATO has only achieved a modest effect in pancreatic ductal adenocarcinoma, suggesting that the existing radical scavenging proteins, such as hypoxia inducible factor-1, attenuate the effect. The goal of this study is to investigate the effect of combination treatment of ATO plus PX-478 (hypoxia-inducible factor-1 inhibitor) and its underlying mechanism. Here, we showed that PX-478 robustly strengthened the anti-growth and pro-apoptosis effect of ATO on Panc-1 and BxPC-3 pancreatic cancer cells in vitro. Meanwhile, in vivo mouse xenograft models also showed the synergistic effect of ATO plus PX-478 compared with any single agent. Further studies showed that the anti-tumor effect of ATO plus PX-478 was derived from the reactive oxygen species-induced apoptosis. We next confirmed that Hypoxia-inducible factor-1 cleared reactive oxygen species by its downstream target, forkhead box O transcription factors, and this effect may justify the strategy of ATO plus PX-478 in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Mingxiao Lang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiuchao Wang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hongwei Wang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jie Dong
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - Chungen Lan
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jihui Hao
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chongbiao Huang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xin Li
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ming Yu
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yanhui Yang
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Shengyu Yang
- Department of Tumor Biology and Comprehensive Melanoma Research Center, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - He Ren
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| |
Collapse
|
99
|
Takahashi N, Davy PMC, Gardner LH, Mathews J, Yamazaki Y, Allsopp RC. Hypoxia Inducible Factor 1 Alpha Is Expressed in Germ Cells throughout the Murine Life Cycle. PLoS One 2016; 11:e0154309. [PMID: 27148974 PMCID: PMC4858237 DOI: 10.1371/journal.pone.0154309] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 04/12/2016] [Indexed: 01/09/2023] Open
Abstract
Pluripotent stem cells of the early embryo, and germ line cells, are essential to ensure uncompromised development to adulthood as well as species propagation, respectively. Recently, the transcription factor hypoxia inducible factor 1 alpha (Hif1α) has been shown to have important roles in embryonic stem cells; in particular, regulation of conversion to glycolytic metabolism and, as we have shown, maintenance of functional levels of telomerase. In the present study, we sought to assess whether Hif1α was also expressed in the primitive cells of the murine embryo. We observed expression of Hif1α in pre-implantation embryos, specifically the 2-cell stage, morula, and blastocyst. Robust Hif1α expression was also observed in male and female primordial germ cells. We subsequently assessed whether Hif1α was expressed in adult male and female germ cells. In the testis, Hif1α was robustly expressed in spermatogonial cells, in both juvenile (6-week old) and adult (3-month old) males. In the ovaries, Hif1α was expressed in mature oocytes from adult females, as assessed both in situ and in individual oocytes flushed from super-ovulated females. Analysis of Hif1α transcript levels indicates a mechanism of regulation during early development that involves stockpiling of Hif1α protein in mature oocytes, presumably to provide protection from hypoxic stress until the gene is re-activated at the blastocyst stage. Together, these observations show that Hif1α is expressed throughout the life-cycle, including both the male and female germ line, and point to an important role for Hif1α in early progenitor cells.
Collapse
Affiliation(s)
- Natsumi Takahashi
- Institute for Biogenesis Research, John A Burns School of Medicine, Honolulu, Hawaii, United States of America
| | - Philip M. C. Davy
- Institute for Biogenesis Research, John A Burns School of Medicine, Honolulu, Hawaii, United States of America
| | - Lauren H. Gardner
- Institute for Biogenesis Research, John A Burns School of Medicine, Honolulu, Hawaii, United States of America
| | - Juanita Mathews
- Institute for Biogenesis Research, John A Burns School of Medicine, Honolulu, Hawaii, United States of America
| | - Yuki Yamazaki
- Institute for Biogenesis Research, John A Burns School of Medicine, Honolulu, Hawaii, United States of America
| | - Richard C. Allsopp
- Institute for Biogenesis Research, John A Burns School of Medicine, Honolulu, Hawaii, United States of America
- * E-mail:
| |
Collapse
|
100
|
Characterization of temporal expressions of FOXO and pFOXO proteins in the hippocampus by kainic acid in mice: involvement of NMDA and non-NMDA receptors. Arch Pharm Res 2016; 39:660-7. [PMID: 26987339 DOI: 10.1007/s12272-016-0733-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 03/12/2016] [Indexed: 10/22/2022]
Abstract
In the present study, we characterized the expression and role of forkhead box O (FoxO3a) in kainic acid (KA)-induced hippocampal neuronal cell death. FoxO3a and pFoxO3a expression in the CA1, CA2, and dentate gyrus regions in the hippocampus increased 0.5 and 1 h after intracerebroventricular administration of KA. In addition, both FoxO3a and pFoxO3a expression in the hippocampal CA3 region increased significantly and equally for 1 h but decreased gradually for 24 h after KA administration. In particular, the KA-induced increases in FoxO3a and pFoxO3a expression in the hippocampal CA3 region were inhibited by pretreatment with the N-methyl-D-aspartate (NMDA) receptor antagonist (MK-801, dizocilpine, 1 µg/5 µl) or a non-NMDA receptor antagonist (CNQX, 6-cyano-7-nitroquinoxaline-2,3-dione, 0.5 µg/5 µl). Furthermore, dizocilpine and CNQX produced a neuroprotective effect against KA-induced neuronal death in the CA3 region of the hippocampus. Our results suggest that FoxO3a and pFoxO3 expression is upregulated by KA. Both FoxO3a and pFoxO3a expression appear to be responsible for KA-induced neuronal death in the CA3 region of the hippocampus.
Collapse
|