1
|
Li M, Tian X, Li X, Huang M, Huang S, Wu Y, Jiang M, Shi Y, Shi L, Wang Z. Diverse energy metabolism patterns in females in Neodon fuscus, Lasiopodomys brandtii, and Mus musculus revealed by comparative transcriptomics under hypoxic conditions. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 783:147130. [PMID: 34088150 DOI: 10.1016/j.scitotenv.2021.147130] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 03/28/2021] [Accepted: 04/09/2021] [Indexed: 06/12/2023]
Abstract
The effects of global warming and anthropogenic disturbance force animals to migrate from lower to higher elevations to find suitable new habitats. As such migrations increase hypoxic stress on the animals, it is important to understand how plateau- and plain-dwelling animals respond to low-oxygen environments. We used comparative transcriptomics to explore the response of Neodon fuscus, Lasiopodomys brandtii, and Mus musculus skeletal muscle tissues to hypoxic conditions. Results indicate that these species have adopted different oxygen transport and energy metabolism strategies for dealing with a hypoxic environment. N. fuscus promotes oxygen transport by increasing hemoglobin synthesis and reduces the risk of thrombosis through cooperative regulation of genes, including Fga, Fgb, Alb, and Ttr; genes such as Acs16, Gpat4, and Ndufb7 are involved in regulating lipid synthesis, fatty acid β-oxidation, hemoglobin synthesis, and electron-linked transmission, thereby maintaining a normal energy supply in hypoxic conditions. In contrast, the oxygen-carrying capacity and angiogenesis of red blood cells in L. brandtii are promoted by genes in the CYP and COL families; this species maintains its bodily energy supply by enhancing the pentose phosphate pathway and mitochondrial fatty acid synthesis pathway. However, under hypoxia, M. musculus cannot effectively transport additional oxygen; thus, its cell cycle, proliferation, and migration are somewhat affected. Given its lack of hypoxic tolerance experience, M. musculus also shows significantly reduced oxidative phosphorylation levels under hypoxic conditions. Our results suggest that the glucose capacity of M. musculus skeletal muscle does not provide sufficient energy during hypoxia; thus, we hypothesize that it supplements its bodily energy by synthesizing ketone bodies. For the first time, we describe the energy metabolism pathways of N. fuscus and L. brandtii skeletal muscle tissues under hypoxic conditions. Our findings, therefore, improve our understanding of how vertebrates thrive in high altitude and plain habitats when faced with hypoxic conditions.
Collapse
Affiliation(s)
- Mengyang Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Xiangyu Tian
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Xiujuan Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Maolin Huang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Shuang Huang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yue Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Mengwan Jiang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yuhua Shi
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Luye Shi
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China.
| | - Zhenlong Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; School of Physical Education (Main campus), Zhengzhou University, Zhengzhou 450001, Henan, China.
| |
Collapse
|
2
|
Sadhu N, Jhun EH, Posen A, Yao Y, He Y, Molokie RE, Wilkie DJ, Wang ZJ. Phenylethanolamine N-methyltransferase gene polymorphisms associate with crisis pain in sickle cell disease patients. Pharmacogenomics 2020; 21:269-278. [PMID: 32162598 DOI: 10.2217/pgs-2019-0096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Phenylethanolamine N-methyltransferase (PNMT) catalyzes the conversion of sympathetic neurotransmitter norepinephrine to epinephrine. We examined the association of PNMT polymorphisms with acute and chronic pain in sickle cell disease (SCD). Methods: Utilization of emergency care owing to painful crisis was used as a marker for acute pain in 131 patients with SCD. Results: rs876493 A allele, rs2934965 T allele and rs2941523 G allele were significantly associated with decreased utilization (p ≤ 0.05). rs876493 A allele showed association with utilization in females (p = 0.003), not males (p = 0.803). rs2934965 T allele and rs2941523 G allele were predicted to cause loss of putative transcription factor binding sites. This is the first report of the association of PNMT polymorphisms with acute crisis pain in SCD. Together with our previous findings in catechol-o-methyltransferase, polymorphisms in catecholamine metabolizing enzymes appear to primarily influence acute pain in SCD.
Collapse
Affiliation(s)
- Nilanjana Sadhu
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60607, USA
| | - Ellie H Jhun
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60607, USA
| | - Andrew Posen
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60607, USA
| | - Yingwei Yao
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, FL, USA
| | - Ying He
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60607, USA.,Comprehensive Sickle Cell Center, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Robert E Molokie
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60607, USA.,Comprehensive Sickle Cell Center, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Jesse Brown Veteran's Administration Medical Center, Chicago, IL, USA.,Division of Hematology/Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Diana J Wilkie
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, FL, USA
| | - Zaijie J Wang
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60607, USA.,Comprehensive Sickle Cell Center, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
3
|
Du Y, Yang X, Gong Q, Xu Z, Cheng Y, Su G. Inhibitor of growth 4 affects hypoxia-induced migration and angiogenesis regulation in retinal pigment epithelial cells. J Cell Physiol 2019; 234:15243-15256. [PMID: 30667053 DOI: 10.1002/jcp.28170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Inhibitor of growth 4 (ING4), a potential tumor suppressor, is implicated in cell migration and angiogenesis. However, its effects on diabetic retinopathy (DR) have not been elucidated. In this study, we aimed to evaluate ING4 expression in normal and diabetic rats and clarify its effects on hypoxia-induced dysfunction in human retinal pigment epithelial (ARPE-19) cells. A Type 1 diabetic model was generated by injecting rats intraperitoneally with streptozotocin and then killed them 4, 8, or 12 weeks later. ING4 expression in retinal tissue was detected using western blot analysis, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), and immunohistochemistry assays. After transfection with an ING4 overexpression lentiviral vector or small interfering RNA (siRNA), ARPE-19 migration under hypoxia was tested using wound healing and transwell assays. The angiogenic effect of conditioned medium (CM) from ARPE-19 cells was examined by assessing human retinal endothelial cell (HREC) capillary tube formation. Additionally, western blot analysis and RT-qPCR were performed to investigate the signaling pathways in which ING4, specificity protein 1 (Sp1), matrix metalloproteinase 2 (MMP-2), MMP-9, and vascular endothelial growth factor A (VEGF-A) were involved. Here, we found that ING4 expression was significantly reduced in the diabetic rats' retinal tissue. Silencing ING4 aggravated hypoxia-induced ARPE-19 cell migration. CM collected from ING4 siRNA-transfected ARPE-19 cells under hypoxia promoted HREC angiogenesis. These effects were reversed by ING4 overexpression. Furthermore, ING4 suppressed MMP-2, MMP-9, and VEGF-A expression in an Sp1-dependent manner in hypoxia-conditioned ARPE-19 cells. Overall, our results provide valuable mechanistic insights into the protective effects of ING4 on hypoxia-induced migration and angiogenesis regulation in ARPE-19 cells. Restoring ING4 may be a novel strategy for treating DR.
Collapse
Affiliation(s)
- Yang Du
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Xinyue Yang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Qiaoyun Gong
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Zhixiang Xu
- Department of Medicine, Division of Hematology/Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Yan Cheng
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Guanfang Su
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
4
|
Morozova AY, Arutjunyan AV, Milyutina YP, Morozova PY, Kozina LS, Zhuravin IA. The Dynamics of the Contents of Neurotrophic Factors in Early Ontogeny in the Brain Structures of Rats Subjected to Prenatal Hypoxia. NEUROCHEM J+ 2018. [DOI: 10.1134/s181971241803008x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
5
|
Phenylethanolamine N-methyltransferase gene expression in PC12 cells exposed to intermittent hypoxia. Neurosci Lett 2018; 666:169-174. [DOI: 10.1016/j.neulet.2017.12.056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/23/2017] [Accepted: 12/26/2017] [Indexed: 11/22/2022]
|
6
|
Ascenzi P, di Masi A, Leboffe L, Fiocchetti M, Nuzzo MT, Brunori M, Marino M. Neuroglobin: From structure to function in health and disease. Mol Aspects Med 2016; 52:1-48. [DOI: 10.1016/j.mam.2016.10.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 10/27/2016] [Accepted: 10/27/2016] [Indexed: 01/01/2023]
|
7
|
Nguyen P, Khurana S, Peltsch H, Grandbois J, Eibl J, Crispo J, Ansell D, Tai TC. Prenatal glucocorticoid exposure programs adrenal PNMT expression and adult hypertension. J Endocrinol 2015; 227:117-27. [PMID: 26475702 DOI: 10.1530/joe-15-0244] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prenatal exposure to glucocorticoids (GCs) programs for hypertension later in life. The aim of the current study was to examine the impact of prenatal GC exposure on the postnatal regulation of the gene encoding for phenylethanolamine N-methyltransferase (PNMT), the enzyme involved in the biosynthesis of the catecholamine, epinephrine. PNMT has been linked to hypertension and is elevated in animal models of hypertension. Male offspring of Wistar-Kyoto dams treated with dexamethasone (DEX) developed elevated systolic, diastolic and mean arterial blood pressure compared to saline-treated controls. Plasma epinephrine levels were also elevated in adult rats exposed to DEX in utero. RT-PCR analysis revealed adrenal PNMT mRNA was higher in DEX exposed adult rats. This was associated with increased mRNA levels of transcriptional regulators of the PNMT gene: Egr-1, AP-2, and GR. Western blot analyses showed increased expression of PNMT protein, along with increased Egr-1 and GR in adult rats exposed to DEX in utero. Furthermore, gel mobility shift assays showed increased binding of Egr-1 and GR to DNA. These results suggest that increased PNMT gene expression via altered transcriptional activity is a possible mechanism by which prenatal exposure to elevated levels of GCs may program for hypertension later in life.
Collapse
Affiliation(s)
- P Nguyen
- Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada
| | - S Khurana
- Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada
| | - H Peltsch
- Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada
| | - J Grandbois
- Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada
| | - J Eibl
- Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada
| | - J Crispo
- Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada
| | - D Ansell
- Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada
| | - T C Tai
- Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada Medical Sciences DivisionNorthern Ontario School of Medicine, Sudbury, Ontario, CanadaDepartments of BiologyChemistry and BiochemistryBiomolecular Sciences ProgramLaurentian University, Sudbury, Ontario, Canada
| |
Collapse
|
8
|
Fassl A, Tagscherer KE, Richter J, De-Castro Arce J, Savini C, Rösl F, Roth W. Inhibition of Notch1 signaling overcomes resistance to the death ligand Trail by specificity protein 1-dependent upregulation of death receptor 5. Cell Death Dis 2015; 6:e1921. [PMID: 26469969 PMCID: PMC4632291 DOI: 10.1038/cddis.2015.261] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 08/04/2015] [Accepted: 08/05/2015] [Indexed: 11/28/2022]
Abstract
The Notch1 signaling pathway contributes to tumorigenesis by influencing differentiation, proliferation and apoptosis. Here, we demonstrate that inhibition of the Notch1 signaling pathway sensitizes glioblastoma cell lines and glioblastoma initiating cells to apoptosis induced by the death ligand TRAIL. This sensitization occurs through transcriptional upregulation of the death receptor 5 (DR5, TRAIL-R2). The increase in DR5 expression is abrogated by concomitant repression of the transcription factor Sp1, which directly binds to the DR5 promoter in the absence of Notch1 as revealed by chromatin immunoprecipitation. Consistent with these findings, Notch1 inhibition resulted in increased DR5 promoter activity, which was impaired by mutation of one out of two Sp1-binding sites within the proximal DR5 promoter. Moreover, we demonstrate that JNK signaling contributes to the regulation of DR5 expression by Notch1. Taken together, our results identify Notch1 as key driver for TRAIL resistance and suggest Notch1 as a promising target for anti-glioblastoma therapy.
Collapse
Affiliation(s)
- A Fassl
- Molecular Tumor Pathology, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany, and Institute of Pathology, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - K E Tagscherer
- Molecular Tumor Pathology, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany, and Institute of Pathology, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - J Richter
- Molecular Tumor Pathology, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany, and Institute of Pathology, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - J De-Castro Arce
- Division of Viral Transformation Mechanisms, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - C Savini
- Division of Viral Transformation Mechanisms, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - F Rösl
- Division of Viral Transformation Mechanisms, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - W Roth
- Molecular Tumor Pathology, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany, and Institute of Pathology, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| |
Collapse
|
9
|
Meshkova EM, Tomilova IK. Biogenic amines in the fetal and newborn brain under normal conditions and in antenatal hypoxia. NEUROCHEM J+ 2015. [DOI: 10.1134/s1819712415030083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
10
|
Banerjee J, Al-Wadei HAN, Al-Wadei MH, Dagnon K, Schuller HM. Differential modulation of nicotine-induced gemcitabine resistance by GABA receptor agonists in pancreatic cancer cell xenografts and in vitro. BMC Cancer 2014; 14:725. [PMID: 25260978 PMCID: PMC4190427 DOI: 10.1186/1471-2407-14-725] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 09/23/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Pancreatic cancer is frequently resistant to cancer therapeutics. Smoking and alcoholism are risk factors and pancreatic cancer patients often undergo nicotine replacement therapy (NRT) and treatment for alcohol dependence. Based on our report that low dose nicotine within the range of NRT causes gemcitabine resistance in pancreatic cancer, our current study has tested the hypothesis that GABA or the selective GABA-B-R agonist baclofen used to treat alcohol dependence reverse nicotine-induced gemcitabine resistance in pancreatic cancer. METHODS Using mouse xenografts from the gemcitabine--sensitive pancreatic cancer cell line BXPC-3, we tested the effects of GABA and baclofen on nicotine-induced gemcitabine resistance. The levels of cAMP, p-SRC, p-ERK, p-AKT, p-CREB and cleaved caspase-3 in xenograft tissues were determined by ELISA assays. Expression of the two GABA-B receptors, metalloproteinase-2 and 9 and EGR-1 in xenograft tissues was monitored by Western blotting. Mechanistic studies were conducted in vitro, using cell lines BXPC-3 and PANC-1 and included analyses of cAMP production by ELISA assay and Western blots to determine protein expression of GABA-B receptors, metalloproteinase-2 and 9 and EGR-1. RESULTS Our data show that GABA was as effective as gemcitabine and significantly reversed gemcitabine resistance induced by low dose nicotine in xenografts whereas baclofen did not. These effects of GABA were accompanied by decreases in cAMP, p-CREB, p-AKT, p-Src, p-ERK metalloproteinases-9 and -2 and EGR-1 and increases in cleaved caspase-3 in xenografts whereas baclofen had the opposite effects. In vitro exposure of cells to single doses or seven days of nicotine induced the protein expression of MMP-2, MMP-9 and EGR-1 and these responses were blocked by GABA. Baclofen downregulated the protein expression of GABA-B-Rs in xenograft tissues and in cells exposed to baclofen for seven days in vitro. This response was accompanied by inversed baclofen effects from inhibition of cAMP formation after single dose exposures to stimulation of cAMP formation in cells pretreated for seven days. CONCLUSIONS These findings suggest GABA as a promising single agent for the therapy of pancreatic cancer and to overcome nicotine-induced gemcitabine resistance whereas treatment with baclofen may increase gemcitabine resistance.
Collapse
Affiliation(s)
- Jheelam Banerjee
- />Experimental Oncology Laboratory, Department of Biomedical & Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN USA
| | - Hussein AN Al-Wadei
- />Experimental Oncology Laboratory, Department of Biomedical & Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN USA
- />Department of Preventive Medicine, Sana’a University, Sana’a, Yemen
| | - Mohammed H Al-Wadei
- />Experimental Oncology Laboratory, Department of Biomedical & Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN USA
| | - Koami Dagnon
- />Experimental Oncology Laboratory, Department of Biomedical & Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN USA
| | - Hildegard M Schuller
- />Experimental Oncology Laboratory, Department of Biomedical & Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN USA
| |
Collapse
|
11
|
Prabhakar NR, Semenza GL. Adaptive and maladaptive cardiorespiratory responses to continuous and intermittent hypoxia mediated by hypoxia-inducible factors 1 and 2. Physiol Rev 2012; 92:967-1003. [PMID: 22811423 DOI: 10.1152/physrev.00030.2011] [Citation(s) in RCA: 476] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hypoxia is a fundamental stimulus that impacts cells, tissues, organs, and physiological systems. The discovery of hypoxia-inducible factor-1 (HIF-1) and subsequent identification of other members of the HIF family of transcriptional activators has provided insight into the molecular underpinnings of oxygen homeostasis. This review focuses on the mechanisms of HIF activation and their roles in physiological and pathophysiological responses to hypoxia, with an emphasis on the cardiorespiratory systems. HIFs are heterodimers comprised of an O(2)-regulated HIF-1α or HIF-2α subunit and a constitutively expressed HIF-1β subunit. Induction of HIF activity under conditions of reduced O(2) availability requires stabilization of HIF-1α and HIF-2α due to reduced prolyl hydroxylation, dimerization with HIF-1β, and interaction with coactivators due to decreased asparaginyl hydroxylation. Stimuli other than hypoxia, such as nitric oxide and reactive oxygen species, can also activate HIFs. HIF-1 and HIF-2 are essential for acute O(2) sensing by the carotid body, and their coordinated transcriptional activation is critical for physiological adaptations to chronic hypoxia including erythropoiesis, vascularization, metabolic reprogramming, and ventilatory acclimatization. In contrast, intermittent hypoxia, which occurs in association with sleep-disordered breathing, results in an imbalance between HIF-1α and HIF-2α that causes oxidative stress, leading to cardiorespiratory pathology.
Collapse
Affiliation(s)
- Nanduri R Prabhakar
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, Biological Sciences Division, University of Chicago, Chicago, Illinois, USA.
| | | |
Collapse
|
12
|
Inbar D, Cohen-Armon M, Neumann D. Erythropoietin-driven signalling and cell migration mediated by polyADP-ribosylation. Br J Cancer 2012; 107:1317-26. [PMID: 22955851 PMCID: PMC3494439 DOI: 10.1038/bjc.2012.395] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background: Recombinant human erythropoietin (EPO) is the leading biotechnology engineered hormone for treatment of anaemia associated with chronic conditions including kidney failure and cancer. The finding of EPO receptors on cancer cells has raised the concern that in addition to its action in erythropoiesis, EPO may promote tumour cell growth. We questioned whether EPO-induced signalling and consequent malignant cell manifestation is mediated by polyADP-ribosylation. Methods: Erythropoietin-mediated PARP (polyADP-ribose polymerase-1) activation, gene expression and core histone H4 acetylation were examined in UT7 cells, using western blot analysis, RT–PCR and immunofluorescence. Erythropoietin-driven migration of the human breast epithelial cell line MDA-MB-435 was determined by the scratch assay and in migration chambers. Results: We have found that EPO treatment induced PARP activation. Moreover, EPO-driven c-fos and Egr-1 gene expression as well as histone H4 acetylation were mediated via polyADP-ribosylation. Erythropoietin-induced cell migration was blocked by the PARP inhibitor, ABT-888, indicating an essential role for polyADP-ribosylation in this process. Conclusions: We have identified a novel pathway by which EPO-induced gene expression and breast cancer cell migration are regulated by polyADP-ribosylation. This study introduces new possibilities regarding EPO treatment for cancer-associated anaemia where combining systemic EPO treatment with targeted administration of PARP inhibitors to the tumour may allow safe treatment with EPO, minimising its possible undesirable proliferative effects on the tumour.
Collapse
Affiliation(s)
- D Inbar
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Israel
| | | | | |
Collapse
|
13
|
Wong DL, Tai TC, Wong-Faull DC, Claycomb R, Meloni EG, Myers KM, Carlezon WA, Kvetnansky R. Epinephrine: a short- and long-term regulator of stress and development of illness : a potential new role for epinephrine in stress. Cell Mol Neurobiol 2012; 32:737-48. [PMID: 22090159 PMCID: PMC11498570 DOI: 10.1007/s10571-011-9768-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 10/31/2011] [Indexed: 01/24/2023]
Abstract
Epinephrine (Epi), which initiates short-term responses to cope with stress, is, in part, stress-regulated via genetic control of its biosynthetic enzyme, phenylethanolamine N-methyltransferase (PNMT). In rats, immobilization (IMMO) stress activates the PNMT gene in the adrenal medulla via Egr-1 and Sp1 induction. Yet, elevated Epi induced by acute and chronic stress is associated with stress induced, chronic illnesses of cardiovascular, immune, cancerous, and behavioral etiologies. Major sources of Epi include the adrenal medulla and brainstem. Although catecholamines do not cross the blood-brain barrier, circulating Epi from the adrenal medulla may communicate with the central nervous system and stress circuitry by activating vagal nerve β-adrenergic receptors to release norepinephrine, which could then stimulate release of the same from the nucleus tractus solitarius and locus coeruleus. In turn, the basal lateral amygdala (BLA) may activate to stimulate afferents to the hypothalamus, neocortex, hippocampus, caudate nucleus, and other brain regions sequentially. Recently, we have shown that repeated IMMO or force swim stress may evoke stress resiliency, as suggested by changes in expression and extinction of fear memory in the fear-potentiated startle paradigm. However, concomitant adrenergic changes seem stressor dependent. Present studies aim to identify stressful conditions that elicit stress resiliency versus stress sensitivity, with the goal of developing a model to investigate the potential role of Epi in stress-associated illness. If chronic Epi over expression does elicit illness, possibilities for alternative therapeutics exist through regulating stress-induced Epi expression, adrenergic receptor function and/or corticosteroid effects on Epi, adrenergic receptors and the stress axis.
Collapse
Affiliation(s)
- Dona Lee Wong
- Department of Psychiatry, Harvard Medical School, MA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Transcriptional regulation mechanisms of hypoxia-induced neuroglobin gene expression. Biochem J 2012; 443:153-64. [PMID: 22239089 DOI: 10.1042/bj20111856] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Ngb (neuroglobin) has been identified as a novel endogenous neuroprotectant. However, little is known about the regulatory mechanisms of Ngb expression, especially under conditions of hypoxia. In the present study, we located the core proximal promoter of the mouse Ngb gene to a 554 bp segment, which harbours putative conserved NF-κB (nuclear factor κB)- and Egr1 (early growth-response factor 1) -binding sites. Overexpression and knockdown of transcription factors p65, p50, Egr1 or Sp1 (specificity protein 1) increased and decreased Ngb expression respectively. Experimental assessments with transfections of mutational Ngb gene promoter constructs, as well as EMSA (electrophoretic mobility-shift assay) and ChIP (chromatin immunoprecipitation) assays, demonstrated that NF-κB family members (p65, p50 and cRel), Egr1 and Sp1 bound in vitro and in vivo to the proximal promoter region of the Ngb gene. Moreover, a κB3 site was found as a pivotal cis-element responsible for hypoxia-induced Ngb promoter activity. NF-κB (p65) and Sp1 were also responsible for hypoxia-induced up-regulation of Ngb expression. Although there are no conserved HREs (hypoxia-response elements) in the promoter of the mouse Ngb gene, the results of the present study suggest that HIF-1α (hypoxia-inducible factor-1α) is also involved in hypoxia-induced Ngb up-regulation. In conclusion, we have identified that NF-κB, Egr1 and Sp1 played important roles in the regulation of basal Ngb expression via specific interactions with the mouse Ngb promoter. NF-κB, Sp1 and HIF-1α contributed to the up-regulation of mouse Ngb gene expression under hypoxic conditions.
Collapse
|
15
|
Sequential activation of hypoxia-inducible factor 1 and specificity protein 1 is required for hypoxia-induced transcriptional stimulation of Abcc8. J Cereb Blood Flow Metab 2012; 32:525-36. [PMID: 22086197 PMCID: PMC3293117 DOI: 10.1038/jcbfm.2011.159] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cerebral ischemia causes increased transcription of sulfonylurea receptor 1 (SUR1), which forms SUR1-regulated NC(Ca-ATP) channels linked to cerebral edema. We tested the hypothesis that hypoxia is an initial signal that stimulates transcription of Abcc8, the gene encoding SUR1, via activation of hypoxia-inducible factor 1 (HIF1). In the brain microvascular endothelial cells, hypoxia increased SUR1 abundance and expression of functional SUR1-regulated NC(Ca-ATP) channels. Luciferase reporter activity driven by the Abcc8 promoter was increased by hypoxia and by coexpression of HIF1α. Surprisingly, a series of luciferase reporter assays studying the Abcc8 promoter revealed that binding sites for specificity protein 1 (Sp1), but not for HIF, were required for stimulation of Abcc8 transcription by HIF1α. Luciferase reporter assays studying Sp1 promoters of three species, and chromatin immunoprecipitation analysis in rats after cerebral ischemia, indicated that HIF binds to HIF-binding sites on the Sp1 promoter to stimulate transcription of the Sp1 gene. We conclude that sequential activation of two transcription factors, HIF and Sp1, is required to stimulate transcription of Abcc8 following cerebral ischemia. Sequential gene activation in cerebral ischemia provides a plausible molecular explanation for the prolonged treatment window observed for inhibition of the end-target gene product, SUR1, by glibenclamide.
Collapse
|
16
|
Role of reactive oxygen species in the neural and hormonal regulation of the PNMT gene in PC12 cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2011; 2011:756938. [PMID: 22007271 PMCID: PMC3189585 DOI: 10.1155/2011/756938] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 07/12/2011] [Indexed: 01/10/2023]
Abstract
The stress hormone, epinephrine, is produced predominantly by adrenal chromaffin cells and its biosynthesis is regulated by the enzyme phenylethanolamine N-methyltransferase (PNMT). Studies have demonstrated that PNMT may be regulated hormonally via the hypothalamic-pituitary-adrenal axis and neurally via the stimulation of the splanchnic nerve. Additionally, hypoxia has been shown to play a key role in the regulation of PNMT. The purpose of this study was to examine the impact of reactive oxygen species (ROS) produced by the hypoxia mimetic agent CoCl2, on the hormonal and neural stimulation of PNMT in an in vitro cell culture model, utilizing the rat pheochromocytoma (PC12) cell line. RT-PCR analyses show inductions of the PNMT intron-retaining and intronless mRNA splice variants by CoCl2 (3.0- and 1.76-fold, respectively). Transient transfection assays of cells treated simultaneously with CoCl2 and the synthetic glucocorticoid, dexamethasone, show increased promoter activity (18.5-fold), while mRNA levels of both splice variants do not demonstrate synergistic effects. Similar results were observed when investigating the effects of CoCl2-induced ROS on the neural stimulation of PNMT via forskolin. Our findings demonstrate that CoCl2-induced ROS have synergistic effects on hormonal and neural activation of the PNMT promoter.
Collapse
|
17
|
Wong DL, Tai TC, Wong-Faull DC, Claycomb R, Siddall BJ, Bell RA, Kvetnansky R. Stress and adrenergic function: HIF1α, a potential regulatory switch. Cell Mol Neurobiol 2010; 30:1451-7. [PMID: 21046459 PMCID: PMC11498814 DOI: 10.1007/s10571-010-9567-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Accepted: 09/02/2010] [Indexed: 10/18/2022]
Abstract
Stress elicits adrenal epinephrine and cortisol release into the bloodstream to initiate physiological and behavioral responses to counter and overcome stress, the classic "fight or flight" response (Cannon and De La Paz, Am J Physiol 28:64-70, 1911). Stress and the stress hormone epinephrine also contribute to the pathophysiology of illness, e.g., behavioral disorders, cardiovascular disease, and immune dysfunction. Epinephrine itself is regulated by stress through its biosynthesis by phenylethanolamine N-methyltransferase (PNMT, EC 2.1.1.28). Single and repeated immobilization (IMMO) stress in rats stimulates adrenal PNMT mRNA and protein expression via the transcription factors, Egr-1 and Sp1. Moderate hypoxic stress increases PNMT promoter-driven gene expression and endogenous PNMT mRNA and protein in PC12 cells. Induction is initiated through cAMP and PLC signaling, with PKA, PKC, PI3K, ERK1/2 MAPK, and p38 MAPK continuing downstream signal transduction, followed by activation of HIF1α, Egr-1, and Sp1. While functional Egr-1 and Sp1 binding sites exist within the proximal PNMT promoter, a putative hypoxia response element is a weak HIF binding site. Yet, HIF1α overexpression increases PNMT promoter-driven luciferase activity and endogenous PNMT. When the Egr-1 or Sp1 sites are mutated, HIF1α does not stimulate the PNMT promoter. siRNA knock down of Egr-1 or Sp1 prevents promoter activation while siRNA knock down of HIF1α inhibits Egr-1 and Sp1 induction. Findings suggest that hypoxia activates the PNMT gene indirectly via HIF1α stimulation of Egr-1 and Sp1. Thus, for stress-induced illnesses where adrenergic dysfunction is implicated, HIF1α may be an "on-off" switch regulating adrenergic responses to stress and a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Dona Lee Wong
- Department of Psychiatry, Harvard Medical School, Laboratory of Molecular and Developmental Neurobiology, McLean Hospital, 115 Mill Street, MRC Rm 116, Mail Stop 144, Belmont, MA 02478, USA.
| | | | | | | | | | | | | |
Collapse
|