1
|
Oliveira FRMB, Sousa Soares E, Pillmann Ramos H, Lättig-Tünnemann G, Harms C, Cimarosti H, Sordi R. Renal protection after hemorrhagic shock in rats: Possible involvement of SUMOylation. Biochem Pharmacol 2024; 227:116425. [PMID: 39004233 DOI: 10.1016/j.bcp.2024.116425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Hemorrhagic shock (HS), a leading cause of preventable death, is characterized by severe blood loss and inadequate tissue perfusion. Reoxygenation of ischemic tissues exacerbates organ damage through ischemia-reperfusion injury. SUMOylation has been shown to protect neurons after stroke and is upregulated in response to cellular stress. However, the role of SUMOylation in organ protection after HS is unknown. This study aimed to investigate SUMOylation-mediated organ protection following HS. Male Wistar rats were subjected to HS (blood pressure of 40 ± 2 mmHg, for 90 min) followed by reperfusion. Blood, kidney, and liver samples were collected at various time points after reperfusion to assess organ damage and investigate the profile of SUMO1 and SUMO2/3 conjugation. In addition, human kidney cells (HK-2), treated with the SUMOylation inhibitor TAK-981 or overexpressing SUMO proteins, were subjected to oxygen and glucose deprivation to investigate the role of SUMOylation in hypoxia/reoxygenation injury. The animals presented progressive multiorgan dysfunction, except for the renal system, which showed improvement over time. Compared to the liver, the kidneys displayed distinct patterns in terms of oxidative stress, apoptosis activation, and tissue damage. The global level of SUMO2/3 in renal tissue was also distinct, suggesting a differential role. Pharmacological inhibition of SUMOylation reduced cell viability after hypoxia-reoxygenation damage, while overexpression of SUMO1 or SUMO2 protected the cells. These findings suggest that SUMOylation might play a critical role in cellular protection during ischemia-reperfusion injury in the kidneys, a role not observed in the liver. This difference potentially explains the renal resilience observed in HS animals when compared to other systems.
Collapse
Affiliation(s)
- Filipe Rodolfo Moreira Borges Oliveira
- Department of Pharmacology, Biological Sciences Center, Universidade Federal de Santa Catarina (UFSC), SC, Brazil; Graduate Program in Pharmacology, UFSC, SC, Brazil
| | - Ericks Sousa Soares
- Department of Pharmacology, Biological Sciences Center, Universidade Federal de Santa Catarina (UFSC), SC, Brazil; Graduate Program in Pharmacology, UFSC, SC, Brazil
| | - Hanna Pillmann Ramos
- Department of Pharmacology, Biological Sciences Center, Universidade Federal de Santa Catarina (UFSC), SC, Brazil
| | - Gisela Lättig-Tünnemann
- Klinik und Hochschulambulanz für Neurologie mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Christoph Harms
- Klinik und Hochschulambulanz für Neurologie mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; Centre for Stroke Research, Berlin, Germany; Charité-Universitätsmedizin Berlin, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), partner site Berlin, Germany; Einstein Centre for Neuroscience, Berlin, Germany
| | - Helena Cimarosti
- Department of Pharmacology, Biological Sciences Center, Universidade Federal de Santa Catarina (UFSC), SC, Brazil; Graduate Program in Pharmacology, UFSC, SC, Brazil; Graduate Program in Neuroscience, UFSC, SC, Brazil
| | - Regina Sordi
- Department of Pharmacology, Biological Sciences Center, Universidade Federal de Santa Catarina (UFSC), SC, Brazil; Graduate Program in Pharmacology, UFSC, SC, Brazil.
| |
Collapse
|
2
|
Khongkla E, Uppakara K, Boonmuen N, Bhukhai K, Saengsawang W. A Novel Methodology Using Dexamethasone to Induce Neuronal Differentiation in the CNS-Derived Catecholaminergic CAD Cells. Cell Mol Neurobiol 2022; 42:2337-2353. [PMID: 34059943 PMCID: PMC11421615 DOI: 10.1007/s10571-021-01109-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
The Cath.a-differentiated (CAD) cell line is a central nervous system-derived catecholaminergic cell line originating from tyrosine hydroxylase (TH)-producing neurons located around the locus coeruleus area of the mouse brain. CAD cells have been used as an in vitro model for cellular and molecular studies due to their ability to differentiate under serum-free media conditions. However, the lack of serum-derived survival factors, limits the longevity for differentiated CAD cells to be maintained in healthy conditions; thereby, limiting their use in long-term culture studies. Here, we present a novel differentiation method that utilizes dexamethasone (Dex), a synthetic glucocorticoid receptor agonist. Specifically, we discovered that the addition of 100 µM of Dex into the 1% fetal bovine serum (FBS)-supplemented media effectively induced neuronal differentiation of CAD cells, as characterized by neurite formation and elongation. Dex-differentiated CAD cells exited the cell cycle, stopped proliferating, extended the neurites, and expressed neuronal markers. These effects were dependent on the glucocorticoid receptors (GR) as they were abolished by GR knockdown. Importantly, Dex-differentiated CAD cells showed longer survival duration than serum-free differentiated CAD cells. In addition, RNA-sequencing and qPCR data demonstrate that several genes involved in proliferation, neuronal differentiation, and survival pathways were differentially expressed in the Dex-differentiated cells. This is the first study to reveal Dex as a novel differentiation methodology used to generate postmitotic neuronal CAD cells, which may be utilized as an in vitro neuronal model for cellular and molecular neurobiology research.
Collapse
Affiliation(s)
- Ekkaphot Khongkla
- Department of Physiology, Faculty of Science, Mahidol University, 272 Rama 6 Rd. Payathai, Ratchathewi district, Bangkok, 10400, Thailand
| | - Kwanchanok Uppakara
- Toxicology Graduate Program, Faculty of Sciene, Mahidol University, Bangkok, Thailand
| | - Nittaya Boonmuen
- Department of Physiology, Faculty of Science, Mahidol University, 272 Rama 6 Rd. Payathai, Ratchathewi district, Bangkok, 10400, Thailand
| | - Kanit Bhukhai
- Department of Physiology, Faculty of Science, Mahidol University, 272 Rama 6 Rd. Payathai, Ratchathewi district, Bangkok, 10400, Thailand
| | - Witchuda Saengsawang
- Department of Physiology, Faculty of Science, Mahidol University, 272 Rama 6 Rd. Payathai, Ratchathewi district, Bangkok, 10400, Thailand.
- Faculty of Science, Center of Neuroscience, Mahidol University, Bangkok, Thailand.
- Faculty of Science, Excellent Center for Drug Discovery (ECDD), Mahidol University, Bangkok, Thailand.
| |
Collapse
|
3
|
Sorteberg AL, Halipi V, Wickström M, Shirazi Fard S. The cyclin dependent kinase inhibitor p21Cip1/Waf1 is a therapeutic target in high-risk neuroblastoma. Front Oncol 2022; 12:906194. [PMID: 36147919 PMCID: PMC9486206 DOI: 10.3389/fonc.2022.906194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/02/2022] [Indexed: 11/29/2022] Open
Abstract
Platinum-based chemotherapies such as cisplatin are used as first-line treatment for the paediatric tumour neuroblastoma. Although the majority of neuroblastoma tumours respond to therapy, there is a high fraction of high-risk neuroblastoma patients that eventually relapse with increased resistance. Here, we show that one key determinant of cisplatin sensitivity is phosphorylation of the cyclin-dependent kinase inhibitor p21Cip1/Waf1. A panel of eight neuroblastoma cell lines and a TH-MYCN mouse model were investigated for the expression of p21Cip1/Waf1 using RT-qPCR, Western blot, and immunofluorescence. This was followed by investigation of sensitivity towards cisplatin and the p21Cip1/Waf1 inhibitor UC2288. Whereas the cell lines and the mouse model showed low levels of un-phosphorylated p21Cip1/Waf1, the phosphorylated p21Cip1/Waf1 (Thr145) was highly expressed, which in the cell lines correlated to cisplatin resistance. Furthermore, the neuroblastoma cell lines showed high sensitivity to UC2288, and combination treatment with cisplatin resulted in considerably decreased cell viability and delay in regrowth in the two most resistant cell lines, SK-N-DZ and BE(2)-C. Thus, targeting p21Cip1/Waf1 can offer new treatment strategies and subsequently lead to the design of more efficient combination treatments for high-risk neuroblastoma.
Collapse
|
4
|
Hoffmann CJ, Kuffner MTC, Lips J, Lorenz S, Endres M, Harms C. Zfp580 Regulates Paracrine and Endocrine Igf1 and Igfbp3 Differently in the Brain and Blood After a Murine Stroke. Front Physiol 2022; 13:887180. [PMID: 35557964 PMCID: PMC9089756 DOI: 10.3389/fphys.2022.887180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Insulin-like growth factor 1 (Igf1) and insulin-like growth factor binding protein 3 (Igfbp3) are endocrine and paracrine factors that influence stroke occurrence, severity, and recovery. Low levels of endocrine Igf1 and Igfbp3 were associated with larger infarct volumes and unfavorable outcomes. Paracrine Igf1 is brain cytoprotective and improves functional recovery after stroke. In this study, we evaluated the effects of zinc finger protein 580 (Zfp580) on endocrine and paracrine Igf1 and Igfbp3 after stroke. Zfp580 suppressed the expression of Igf1 and Igfbp3 in cerebral microvascular endothelial cells (bEnd.3) as determined by real-time RT-PCR. Zfp580 was suppressed by combined oxygen and glucose deprivation (OGD) and mediated the effect of OGD on Igf1 and Igfbp3. In vivo, we evaluated paracrine regulation by real-time RT-PCR of brain lysates and endocrine regulation by ELISA of blood samples. Genomic ablation of Zfp580 did not alter basal paracrine or endocrine Igf1 and Igfbp3 levels. After transient middle cerebral artery occlusion (MCAo), Zfp580 was globally elevated in the brain for up to 3 days. Paracrine Igf1 and Igfbp3 were selectively induced in the ischemic hemisphere from day 2 to day 3 or day 1 to day 7, respectively. In Zfp580 knockout mice, the paracrine regulations of Igf1 and Igfbp3 were attenuated while endocrine Igf1 and the molar Igf1/Igfbp3 ratio were increased. In conclusion, Zfp580 differentially controls paracrine and endocrine Igf1 and Igfbp3 after stroke. Inhibition of Zfp580 might be a new treatment target leading to increased activity of Igf1 to improve stroke outcome.
Collapse
Affiliation(s)
- Christian J Hoffmann
- Klinik und Hochschulambulanz Für Neurologie Mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany.,Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Melanie T C Kuffner
- Klinik und Hochschulambulanz Für Neurologie Mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Janet Lips
- Klinik und Hochschulambulanz Für Neurologie Mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Stephanie Lorenz
- Klinik und Hochschulambulanz Für Neurologie Mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Matthias Endres
- Klinik und Hochschulambulanz Für Neurologie Mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany.,Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.,Einstein Center for Neuroscience, Berlin, Germany
| | - Christoph Harms
- Klinik und Hochschulambulanz Für Neurologie Mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany.,Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Einstein Center for Neuroscience, Berlin, Germany
| |
Collapse
|
5
|
Pharmacological intervention of histone deacetylase enzymes in the neurodegenerative disorders. Life Sci 2020; 243:117278. [PMID: 31926248 DOI: 10.1016/j.lfs.2020.117278] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 12/31/2019] [Accepted: 01/01/2020] [Indexed: 02/06/2023]
Abstract
Reversal of aging symptoms and related disorders are the challenging task where epigenetic is a crucial player that includes DNA methylation, histone modification; chromatin remodeling and regulation that are linked to the progression of various neurodegenerative disorders (NDDs). Overexpression of various histone deacetylase (HDACs) can activate Glycogen synthase kinase 3 which promotes the hyperphosphorylation of tau and inhibits its degradation. While HDAC is important for maintaining the neuronal morphology and brain homeostasis, at the same time, these enzymes are promoting neurodegeneration, if it is deregulated. Different experimental models have also confirmed the neuroprotective effects caused by HDAC enzymes through the regulation of neuronal apoptosis, inflammatory response, DNA damage, cell cycle regulation, and metabolic dysfunction. Apart from transcriptional regulation, protein-protein interaction, histone post-translational modifications, deacetylation mechanism of non-histone protein and direct association with disease proteins have been linked to neuronal imbalance. Histone deacetylases inhibitors (HDACi) can be able to alter gene expression and shown its efficacy on experimental models, and in clinical trials for NDD's and found to be a very promising therapeutic agent with certain limitation, for instance, non-specific target effect, isoform-selectivity, specificity, and limited number of predicted biomarkers. Herein, we discussed (i) the catalytic mechanism of the deacetylation process of various HDAC's in in vivo and in vitro experimental models, (ii) how HDACs are participating in neuroprotection as well as in neurodegeneration, (iii) a comprehensive role of HDACi in maintaining neuronal homeostasis and (iv) therapeutic role of biomolecules to modulate HDACs.
Collapse
|
6
|
Xia P, Liu Y, Chen J, Cheng Z. Cell Cycle Proteins as Key Regulators of Postmitotic Cell Death. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2019; 92:641-650. [PMID: 31866779 PMCID: PMC6913832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Cell cycle progression in dividing cells, characterized by faithful replication of the genomic materials and duplication of the original cell, is fundamental for growth and reproduction of all mammalian organisms. Functional maturation of postmitotic cells, however, requires cell cycle exit and terminal differentiation. In mature postmitotic cells, many cell cycle proteins remain to be expressed, or can be induced and reactivated in pathological conditions such as traumatic injury and degenerative diseases. Interestingly, elevated levels of cell cycle proteins in postmitotic cells often do not induce proliferation, but result in aberrant cell cycle reentry and cell death. At present, the cell cycle machinery is known predominantly for regulating cell cycle progression and cell proliferation, albeit accumulating evidence indicates that cell cycle proteins may also control cell death, especially in postmitotic tissues. Herein, we provide a brief summary of these findings and hope to highlight the connection between cell cycle reentry and postmitotic cell death. In addition, we also outline the signaling pathways that have been identified in cell cycle-related cell death. Advanced understanding of the molecular mechanisms underlying cell cycle-related death is of paramount importance because this knowledge can be applied to develop protective strategies against pathologies in postmitotic tissues. Moreover, a full-scope understanding of the cell cycle machinery will allow fine tuning to favor cell proliferation over cell death, thereby potentially promoting tissue regeneration.
Collapse
Affiliation(s)
| | | | | | - Zhaokang Cheng
- To whom all correspondence should be addressed: Zhaokang Cheng, PhD, Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd. Spokane, WA 99202-2131; Tel: 509-358-7741,
| |
Collapse
|
7
|
Dong J, Li J, Li J, Cui L, Meng X, Qu Y, Wang H. The proliferative effect of cortisol on bovine endometrial epithelial cells. Reprod Biol Endocrinol 2019; 17:97. [PMID: 31757215 PMCID: PMC6873581 DOI: 10.1186/s12958-019-0544-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 11/13/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Bovine endometrial epithelial cells (BEECs) undergo regular regeneration after calving. Elevated cortisol concentrations have been reported in postpartum cattle due to various stresses. However, the effects of the physiological level of cortisol on proliferation in BEECs have not been reported. The aim of this study was to investigate whether cortisol can influence the proliferation properties of BEECs and to clarify the possible underlying mechanism. METHODS BEECs were treated with different concentrations of cortisol (5, 15 and 30 ng/mL). The mRNA expression of various growth factors was detected by quantitative reverse transcription-polymerase chain reaction (qPCR), progression of the cell cycle in BEECs was measured using flow cytometric analysis, and the activation of the Wnt/β-catenin and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathways was detected with Western blot and immunofluorescence. RESULTS Cortisol treatment resulted in upregulated mRNA levels of vascular endothelial growth factor (VEGF) and connective tissue growth factor (CTGF); however, it had no influence on transforming growth factor-beta1 (TGF-β1). Cortisol (15 ng/mL) accelerated the cell cycle transition from the G0/G1 to the S phase. Cortisol upregulated the expression of β-catenin, c-Myc, and cyclinD1 and promoted the phosphorylation of PI3K and AKT. CONCLUSIONS These results demonstrated that cortisol may promote proliferation in BEECs by increasing the expression of some growth factors and activating the Wnt/β-catenin and PI3K/AKT signaling pathways.
Collapse
Affiliation(s)
- Junsheng Dong
- grid.268415.cCollege of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 Jiangsu China
| | - Jun Li
- grid.268415.cCollege of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 Jiangsu China
| | - Jianji Li
- grid.268415.cCollege of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 Jiangsu China
| | - Luying Cui
- grid.268415.cCollege of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 Jiangsu China
| | - Xia Meng
- grid.268415.cCollege of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 Jiangsu China
| | - Yang Qu
- grid.268415.cCollege of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 Jiangsu China
| | - Heng Wang
- grid.268415.cCollege of Veterinary Medicine, Yangzhou University, Yangzhou, 225009 Jiangsu China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 Jiangsu China
| |
Collapse
|
8
|
Waetzig V, Haeusgen W, Andres C, Frehse S, Reinecke K, Bruckmueller H, Boehm R, Herdegen T, Cascorbi I. Retinoic acid-induced survival effects in SH-SY5Y neuroblastoma cells. J Cell Biochem 2018; 120:5974-5986. [PMID: 30320919 DOI: 10.1002/jcb.27885] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/19/2018] [Indexed: 12/20/2022]
Abstract
Neuroblastoma is a malignant childhood cancer arising from the embryonic sympathoadrenal lineage of the neural crest. Retinoic acid (RA) is included in the multimodal therapy of patients with high-risk neuroblastoma to eliminate minimal residual disease. However, the formation of RA-resistant cells substantially lowers 5-year overall survival rates. To examine mechanisms that lead to treatment failure, we chose human SH-SY5Y cells, which are known to tolerate incubation with RA by activating the survival kinases Akt and extracellular signal-regulated kinase 1/2. Characterization of downstream pathways showed that both kinases increased the phosphorylation of the ubiquitin ligase mouse double minute homolog 2 (Mdm2) and thereby enhanced p53 degradation. When p53 signaling was sustained by blocking complex formation with Mdm2 or enhancing c-Jun N-terminal kinase (JNK) activation, cell viability was significantly reduced. In addition, Akt-mediated phosphorylation of the cell-cycle regulator p21 stimulated complex formation with caspase-3, which also contributed to cell protection. Thus, treatment with RA augmented survival signaling and attenuated basal apoptotic pathways in SH-SY5Y cells, which increased cell viability.
Collapse
Affiliation(s)
- Vicki Waetzig
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Wiebke Haeusgen
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Cordula Andres
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Sonja Frehse
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Kirstin Reinecke
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Henrike Bruckmueller
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ruwen Boehm
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Thomas Herdegen
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
9
|
Merkouris S, Barde YA, Binley KE, Allen ND, Stepanov AV, Wu NC, Grande G, Lin CW, Li M, Nan X, Chacon-Fernandez P, DiStefano PS, Lindsay RM, Lerner RA, Xie J. Fully human agonist antibodies to TrkB using autocrine cell-based selection from a combinatorial antibody library. Proc Natl Acad Sci U S A 2018; 115:E7023-E7032. [PMID: 29987039 PMCID: PMC6065019 DOI: 10.1073/pnas.1806660115] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The diverse physiological roles of the neurotrophin family have long prompted exploration of their potential as therapeutic agents for nerve injury and neurodegenerative diseases. To date, clinical trials of one family member, brain-derived neurotrophic factor (BDNF), have disappointingly failed to meet desired endpoints. Contributing to these failures is the fact that BDNF is pharmaceutically a nonideal biologic drug candidate. It is a highly charged, yet is a net hydrophobic molecule with a low molecular weight that confers a short t1/2 in man. To circumvent these shortcomings of BDNF as a drug candidate, we have employed a function-based cellular screening assay to select activating antibodies of the BDNF receptor TrkB from a combinatorial human short-chain variable fragment antibody library. We report here the successful selection of several potent TrkB agonist antibodies and detailed biochemical and physiological characterization of one such antibody, ZEB85. By using a human TrkB reporter cell line and BDNF-responsive GABAergic neurons derived from human ES cells, we demonstrate that ZEB85 is a full agonist of TrkB, comparable in potency to BDNF toward human neurons in activation of TrkB phosphorylation, canonical signal transduction, and mRNA transcriptional regulation.
Collapse
Affiliation(s)
- Spyros Merkouris
- School of Biosciences, Cardiff University, CF10 3AX Cardiff, United Kingdom
| | - Yves-Alain Barde
- School of Biosciences, Cardiff University, CF10 3AX Cardiff, United Kingdom
| | - Kate E Binley
- School of Biosciences, Cardiff University, CF10 3AX Cardiff, United Kingdom
| | - Nicholas D Allen
- School of Biosciences, Cardiff University, CF10 3AX Cardiff, United Kingdom
| | - Alexey V Stepanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Nicholas C Wu
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Geramie Grande
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Chih-Wei Lin
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Meng Li
- School of Biosciences, Cardiff University, CF10 3AX Cardiff, United Kingdom
| | - Xinsheng Nan
- School of Biosciences, Cardiff University, CF10 3AX Cardiff, United Kingdom
| | | | | | | | - Richard A Lerner
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037;
| | - Jia Xie
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037;
| |
Collapse
|
10
|
Thomas EA, D'Mello SR. Complex neuroprotective and neurotoxic effects of histone deacetylases. J Neurochem 2018; 145:96-110. [PMID: 29355955 DOI: 10.1111/jnc.14309] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/05/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022]
Abstract
By their ability to shatter quality of life for both patients and caregivers, neurodegenerative diseases are the most devastating of human disorders. Unfortunately, there are no effective or long-terms treatments capable of slowing down the relentless loss of neurons in any of these diseases. One impediment is the lack of detailed knowledge of the molecular mechanisms underlying the processes of neurodegeneration. While some neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, are mostly sporadic in nature, driven by both environment and genetic susceptibility, many others, including Huntington's disease, spinocerebellar ataxias, and spinal-bulbar muscular atrophy, are genetically inherited disorders. Surprisingly, given their different roots and etiologies, both sporadic and genetic neurodegenerative disorders have been linked to disease mechanisms involving histone deacetylase (HDAC) proteins, which consists of 18 family members with diverse functions. While most studies have implicated certain HDAC subtypes in promoting neurodegeneration, a substantial body of literature suggests that other HDAC proteins can preserve neuronal viability. Of particular interest, however, is the recent realization that a single HDAC subtype can have both neuroprotective and neurotoxic effects. Diverse mechanisms, beyond transcriptional regulation have been linked to these effects, including deacetylation of non-histone proteins, protein-protein interactions, post-translational modifications of the HDAC proteins themselves and direct interactions with disease proteins. The roles of these HDACs in both sporadic and genetic neurodegenerative diseases will be discussed in the current review.
Collapse
Affiliation(s)
- Elizabeth A Thomas
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California, USA
| | - Santosh R D'Mello
- Department of Biological Sciences, Southern Methodist University, Dallas, Texas, USA
| |
Collapse
|
11
|
Interaction of ARC and Daxx: A Novel Endogenous Target to Preserve Motor Function and Cell Loss after Focal Brain Ischemia in Mice. J Neurosci 2017; 36:8132-48. [PMID: 27488634 DOI: 10.1523/jneurosci.4428-15.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 06/07/2016] [Indexed: 12/26/2022] Open
Abstract
UNLABELLED The aim of this study was to explore the signaling and neuroprotective effect of transactivator of transcription (TAT) protein transduction of the apoptosis repressor with CARD (ARC) in in vitro and in vivo models of cerebral ischemia in mice. In mice, transient focal cerebral ischemia reduced endogenous ARC protein in neurons in the ischemic striatum at early reperfusion time points, and in primary neuronal cultures, RNA interference resulted in greater neuronal susceptibility to oxygen glucose deprivation (OGD). TAT.ARC protein delivery led to a dose-dependent better survival after OGD. Infarct sizes 72 h after 60 min middle cerebral artery occlusion (MCAo) were on average 30 ± 8% (mean ± SD; p = 0.005; T2-weighted MRI) smaller in TAT.ARC-treated mice (1 μg intraventricularly during MCAo) compared with controls. TAT.ARC-treated mice showed better performance in the pole test compared with TAT.β-Gal-treated controls. Importantly, post-stroke treatment (3 h after MCAo) was still effective in affording reduced lesion volume by 20 ± 7% (mean ± SD; p < 0.05) and better functional outcome compared with controls. Delayed treatment in mice subjected to 30 min MCAo led to sustained neuroprotection and functional behavior benefits for at least 28 d. Functionally, TAT.ARC treatment inhibited DAXX-ASK1-JNK signaling in the ischemic brain. ARC interacts with DAXX in a CARD-dependent manner to block DAXX trafficking and ASK1-JNK activation. Our work identifies for the first time ARC-DAXX binding to block ASK1-JNK activation as an ARC-specific endogenous mechanism that interferes with neuronal cell death and ischemic brain injury. Delayed delivery of TAT.ARC may present a promising target for stroke therapy. SIGNIFICANCE STATEMENT Up to now, the only successful pharmacological target of human ischemic stroke is thrombolysis. Neuroprotective pharmacological strategies are needed to accompany therapies aiming to achieve reperfusion. We describe that apoptosis repressor with CARD (ARC) interacts and inhibits DAXX and proximal signals of cell death. In a murine stroke model mimicking human malignant infarction in the territory of the middle cerebral artery, TAT.ARC salvages brain tissue when given during occlusion or 3 h delayed with sustained functional benefits (28 d). This is a promising novel therapeutic approach because it appears to be effective in a model producing severe injury by interfering with an array of proximal signals and effectors of the ischemic cascade, upstream of JNK, caspases, and BIM and BAX activation.
Collapse
|
12
|
Freyer D, Harms C. Kinetic Lactate Dehydrogenase Assay for Detection of Cell Damage in Primary Neuronal Cell Cultures. Bio Protoc 2017; 7:e2308. [PMID: 34541076 DOI: 10.21769/bioprotoc.2308] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/31/2017] [Accepted: 04/18/2017] [Indexed: 11/02/2022] Open
Abstract
The aim of many in vitro models of acute or chronic degenerative disorders in the neurobiology field is the assessment of survival or damage of neuronal cells. Damage of cells is associated with loss of outer cell membrane integrity and leakage of cytoplasmic cellular proteins. Therefore, activity assays of cytoplasmic enzymes in supernatants of cell cultures serve as a practicable tool for quantification of cellular injury (Koh and Choi, 1987; Bruer et al., 1997 ). Lactate dehydrogenase (LDH) is such a ubiquitously expressed cytosolic enzyme, which is very stable due to a very long protein half-life (Hsieh and Blumenthal, 1956; Koh and Cotman, 1992; Koh et al., 1995 ).
Collapse
Affiliation(s)
- Dorette Freyer
- Department of Experimental Neurology, Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Harms
- Department of Experimental Neurology, Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
13
|
Reduced Expression of Foxp1 as a Contributing Factor in Huntington's Disease. J Neurosci 2017; 37:6575-6587. [PMID: 28550168 DOI: 10.1523/jneurosci.3612-16.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 05/08/2017] [Accepted: 05/11/2017] [Indexed: 12/31/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disease caused by a polyglutamine expansion in the huntington protein (htt). The neuropathological hallmark of HD is the loss of neurons in the striatum and, to a lesser extent, in the cortex. Foxp1 is a member of the Forkhead family of transcription factors expressed selectively in the striatum and the cortex. In the brain, three major Foxp1 isoforms are expressed: isoform-A (∼90 kDa), isoform-D (∼70 kDa), and isoform-C (∼50 kDa). We find that expression of Foxp1 isoform-A and -D is selectively reduced in the striatum and cortex of R6/2 HD mice as well as in the striatum of HD patients. Furthermore, expression of mutant htt in neurons results in the downregulation of Foxp1 Elevating expression of isoform-A or -D protects cortical neurons from death caused by the expression of mutant htt On the other hand, knockdown of Foxp1 promotes death in otherwise healthy neurons. Neuroprotection by Foxp1 is likely to be mediated by the transcriptional stimulation of the cell-cycle inhibitory protein p21Waf1/Cip1 Consistently, Foxp1 activates transcription of the p21Waf1/Cip1 gene promoter, and overexpression of Foxp1 in neurons results in the elevation of p21 expression. Moreover, knocking down of p21Waf1/Cip1 blocks the ability of Foxp1 to protect neurons from mut-Htt-induced neurotoxicity. We propose that the selective vulnerability of neurons of the striatum and cortex in HD is related to the loss of expression of Foxp1, a protein that is highly expressed in these neurons and required for their survival.SIGNIFICANCE STATEMENT Although the mutant huntingtin gene is expressed widely, neurons of the striatum and cortex are selectively affected in Huntington's disease (HD). Our results suggest that this selectivity is attributable to the reduced expression of Foxp1, a protein expressed selectively in striatal and cortical neurons that plays a neuroprotective role in these cells. We show that protection by Foxp1 involves stimulation of the p21Waf1/Cip1 (Cdkn1a) gene. Although three major Foxp1 isoforms (A, C, and D) are expressed in the brain, only isoform-A has been studied in the nervous system. We show that isoform-D is also expressed selectively, neuroprotective and downregulated in HD mice and patients. Our results suggest that Foxp1 might be an attractive therapeutic target for HD.
Collapse
|
14
|
Fielder E, von Zglinicki T, Jurk D. The DNA Damage Response in Neurons: Die by Apoptosis or Survive in a Senescence-Like State? J Alzheimers Dis 2017; 60:S107-S131. [PMID: 28436392 DOI: 10.3233/jad-161221] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurons are exposed to high levels of DNA damage from both physiological and pathological sources. Neurons are post-mitotic and their loss cannot be easily recovered from; to cope with DNA damage a complex pathway called the DNA damage response (DDR) has evolved. This recognizes the damage, and through kinases such as ataxia-telangiectasia mutated (ATM) recruits and activates downstream factors that mediate either apoptosis or survival. This choice between these opposing outcomes integrates many inputs primarily through a number of key cross-road proteins, including ATM, p53, and p21. Evidence of re-entry into the cell-cycle by neurons can be seen in aging and diseases such as Alzheimer's disease. This aberrant cell-cycle re-entry is lethal and can lead to the apoptotic death of the neuron. Many downstream factors of the DDR promote cell-cycle arrest in response to damage and appear to protect neurons from apoptotic death. However, neurons surviving with a persistently activated DDR show all the features known from cell senescence; including metabolic dysregulation, mitochondrial dysfunction, and the hyper-production of pro-oxidant, pro-inflammatory and matrix-remodeling factors. These cells, termed senescence-like neurons, can negatively influence the extracellular environment and may promote induction of the same phenotype in surrounding cells, as well as driving aging and age-related diseases. Recently developed interventions targeting the DDR and/or the senescent phenotype in a range of non-neuronal tissues are being reviewed as they might become of therapeutic interest in neurodegenerative diseases.
Collapse
Affiliation(s)
- Edward Fielder
- The Ageing Biology Centre and Institute for Cell and Molecular Biology, Newcastle University, Newcastle Upon Tyne, UK
| | - Thomas von Zglinicki
- The Ageing Biology Centre and Institute for Cell and Molecular Biology, Newcastle University, Newcastle Upon Tyne, UK
| | - Diana Jurk
- The Ageing Biology Centre and Institute for Cell and Molecular Biology, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
15
|
Simone JJ, McCormick CM. Intracellular signalling and plasma hormone profiles associated with the expression of unconditioned and conditioned fear and anxiety in female rats. Physiol Behav 2016; 169:234-244. [PMID: 27939364 DOI: 10.1016/j.physbeh.2016.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/01/2016] [Accepted: 12/01/2016] [Indexed: 12/11/2022]
Abstract
There is considerable overlap in the neural regions and intracellular signalling pathways implicated in anxiety and fear, although less is known in females. Here, we investigated whether unconditioned and conditioned fear are associated with distinct patterns of expression of extracellular signal-regulated kinase-1 and -2 (ERK1/2), protein kinase B (Akt), and calcineurin (CaN) (proteins that are key regulators of the expression of and/or memory processes of fear and anxiety) in the dorsal and ventral hippocampus, medial prefrontal cortex, and amygdala (important regions in neural fear circuitry) of adult female rats, and used a multivariate approach to find patterns of signalling that might discriminate between the different states of fear. To isolate fear to the conditioned cue from generalized fear to the test context, rats were conditioned to an auditory tone (i.e. tone paired with footshock) and twenty-four hours later exposed to a novel context in the presence or absence of the conditioned cue. A third group that was exposed to the conditioning context without undergoing fear conditioning was included to control for unconditioned responses to the testing procedures, which are anxiogenic. A discriminate function analysis and MANOVA determined that hippocampal signalling best discriminated the three groups from each other. The addition of values for plasma concentrations of corticosterone and progesterone (as indices of activation of the hypothalamic-pituitary-adrenal stress axis) to statistical analyses increased the separation of the three groups. There was high degree of association among the three signalling molecules in the four brain regions within each group. There was an absence of the associations between the medial prefrontal cortex and the amygdala in the cued fear recall group that were strong for the non-conditioned group. These results demonstrated unique neuronal and hormonal signalling profiles associated with unconditioned, generalized, and conditioned fear expression in females and highlight the importance of including appropriate comparisons to best discriminate between these different emotional states.
Collapse
Affiliation(s)
- Jonathan J Simone
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, Canada, L2S 3A1
| | - Cheryl M McCormick
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, Canada, L2S 3A1; Centre for Neuroscience, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, Canada, L2S 3A1; Department of Psychology, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, Ontario, Canada, L2S 3A1.
| |
Collapse
|
16
|
Hauck L, Grothe D, Billia F. p21(CIP1/WAF1)-dependent inhibition of cardiac hypertrophy in response to Angiotensin II involves Akt/Myc and pRb signaling. Peptides 2016; 83:38-48. [PMID: 27486069 DOI: 10.1016/j.peptides.2016.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 07/14/2016] [Accepted: 07/19/2016] [Indexed: 10/21/2022]
Abstract
The cyclin-dependent kinase inhibitor p21(CIP1/WAF1) (p21) is highly expressed in the adult heart. However, in response to stress, its expression is downregulated. Therefore, we investigated the role of p21 in the regulation of cardiac hypertrophic growth. At 2 months of age, p21 knockout mice (p21KO) lack an overt cardiac phenotype. In contrast, by 10 months of age, p21KO developed age-dependent cardiac hypertrophy and heart failure. After 3 weeks of trans-aortic banding (TAB), the heart/body weight ratio in 11 week old p21KO mice increased by 57%, as compared to 42% in wild type mice indicating that p21KO have a higher susceptibility to pressure overload-induced cardiac hypertrophy. We then chronically infused 8 week old wild type mice with Angiotensin II (2.0mg/kg/min) or saline subcutaneously by osmotic pumps for 14 days. Recombinant TAT conjugated p21 protein variants (10mg/kg body weight) or saline were intraperitoneally injected once daily for 14 days into Angiotensin II and saline-infused animals. Angiotensin II treated mice developed pathological cardiac hypertrophy with an average increase of 38% in heart/body weight ratios, as compared to saline-treated controls. Reconstitution of p21 function by TAT.p21 protein transduction prevented Angiotensin II-dependent development of cardiac hypertrophy and failure. Taken together, our genetic and biochemical data show an important function of p21 in the regulation of growth-related processes in the heart.
Collapse
Affiliation(s)
- Ludger Hauck
- Toronto General Research Institute, 100 College St., Toronto, Ontario, M5G 1L7, Canada.
| | - Daniela Grothe
- Toronto General Research Institute, 100 College St., Toronto, Ontario, M5G 1L7, Canada.
| | - Filio Billia
- Toronto General Research Institute, 100 College St., Toronto, Ontario, M5G 1L7, Canada; Division of Cardiology, University Health Network (UHN), 200 Elizabeth St., Toronto, Ontario, M5G 2C4, Canada; Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Canada; Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5G 1A8, Canada.
| |
Collapse
|
17
|
Pfister JA, D'Mello SR. Regulation of Neuronal Survival by Nucleophosmin 1 (NPM1) Is Dependent on Its Expression Level, Subcellular Localization, and Oligomerization Status. J Biol Chem 2016; 291:20787-97. [PMID: 27510036 DOI: 10.1074/jbc.m116.723015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Indexed: 11/06/2022] Open
Abstract
NPM1 (nucleophosmin 1) is a nucleolar phosphoprotein that regulates many cellular processes, including ribosome biogenesis, proliferation, and genomic integrity. Although its role in proliferating cell types and tissues has been extensively investigated, little is known about its function in neurons and in the brain where it is highly expressed. We report that NPM1 protein expression is increased selectively in the striatum in both the R6/2 transgenic and 3-nitropropionic acid-injected mouse models of Huntington's disease. Examination of the effect of ectopic expression on cultured neurons revealed that increasing NPM1 is toxic to otherwise healthy cerebellar granule and cortical neurons. Toxicity is dependent on its cytoplasmic localization and oligomerization status. Forced retention of NPM1 in the nucleus, as well as inhibiting its ability to oligomerize, not only neutralizes NPM1 toxicity but also renders it protective against apoptosis. Although not blocked by pharmacological inhibition of the pro-apoptotic molecules, JNK, glycogen synthase kinase 3 beta, or caspases, toxicity is blocked by compounds targeting cyclin-dependent kinases (CDKs), as well as by dominant-negative forms of CDK1 and CDK2 and the pan-CDK inhibitor, p21(Cip1/Waf1) Although induced in in vivo Huntington's disease models, NPM1 protein levels are unchanged in cultured cerebellar granule and cortical neurons induced to die by low potassium or homocysteic acid treatment, respectively. Moreover, and counterintuitively, knockdown of its expression or inhibition of endogenous NPM1 oligomerization in these cultured neurons is toxic. Taken together, our study suggests that although neurons need NPM1 for survival, an increase in its expression beyond physiological levels and its translocation to the cytoplasm leads to death through abortive cell cycle induction.
Collapse
Affiliation(s)
- Jason A Pfister
- From the Department of Molecular and Cell Biology, University of Texas at Dallas, Richardson, Texas 75080 and
| | - Santosh R D'Mello
- Department of Biological Sciences, Southern Methodist University, Dallas, Texas 75275
| |
Collapse
|
18
|
Du R, Zhou J, Lorenzano S, Liu W, Charoenvimolphan N, Qian B, Xu J, Wang J, Zhang X, Wang X, Berndt A, Devan WJ, Valant VJ, Wang J, Furie KL, Rosand J, Rost N, Friedlander RM, Paigen B, Weiss ST. Integrative Mouse and Human Studies Implicate ANGPT1 and ZBTB7C as Susceptibility Genes to Ischemic Injury. Stroke 2015; 46:3514-22. [PMID: 26542693 DOI: 10.1161/strokeaha.115.010767] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/01/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND PURPOSE The extent of ischemic injury in response to cerebral ischemia is known to be affected by native vasculature. However, the nonvascular and dynamic vascular responses and their genetic basis are not well understood. METHODS We performed a genome-wide association study in 235 mice from 33 inbred strains using the middle cerebral artery occlusion model. Population structure and genetic relatedness were accounted for using the efficient mixed-model association method. Human orthologs to the genes associated with the significant and suggestive single-nucleotide polymorphisms from the mouse strain survey were examined in patients with M1 occlusions admitted with signs and symptoms of acute ischemic stroke. RESULTS We identified 4 genome-wide significant and suggestive single-nucleotide polymorphisms to be associated with infarct volume in mice (rs3694965, P=2.17×10(-7); rs31924033, P=5.61×10(-6); rs32249495, P=2.08×10(-7); and rs3677406, P=9.56×10(-6)). rs32249495, which corresponds to angiopoietin-1 (ANGPT1), was also significant in the recessive model in humans, whereas rs1944577, which corresponds to ZBTB7C, was nominally significant in both the additive and dominant genetic models in humans. ZBTB7C was shown to be upregulated in endothelial cells using both in vitro and in vivo models of ischemia. CONCLUSIONS Genetic variations of ANGPT1 and ZBTB7C are associated with increased infarct size in both mice and humans. ZBTB7C may modulate the ischemic response via neuronal apoptosis and dynamic collateralization and, in addition to ANGPT1, may serve as potential novel targets for treatments of cerebral ischemia.
Collapse
Affiliation(s)
- Rose Du
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.).
| | - Jing Zhou
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Svetlana Lorenzano
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Wenming Liu
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Nareerat Charoenvimolphan
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Baogang Qian
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Jun Xu
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Jian Wang
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Xinmu Zhang
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Xin Wang
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Annerose Berndt
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - William J Devan
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Valerie J Valant
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Jinyi Wang
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Karen L Furie
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Jonathan Rosand
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Natalia Rost
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Robert M Friedlander
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Beverly Paigen
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| | - Scott T Weiss
- From the Department of Neurosurgery (R.D., J.Z., W.L., N.C., B.Q., J.X., J.W., X.Z., X.W.) and Channing Division of Network Medicine, Department of Medicine (R.D., S.T.W.), Brigham and Women's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston (S.L., W.J.D., V.J.V., J.R., N.R.); Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy (S.L.); Department of Chemical Biology, Northwest Agriculture and Forestry University, Shaanxi, People's Republic of China (W.L., J.W.); Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, People's Republic of China (J.X.); The Jackson Laboratory, Bar Harbor, ME (A.B., B.P.); Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.B.) and Department of Neurosurgery (R.M.F.), University of Pittsburgh School of Medicine, PA; Quinnipiac University Frank H. Netter, MD School of Medicine, Hamden, CT (W.J.D.); University of Massachusetts Medical School, Worcester (V.J.V.); and Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (K.L.F.)
| |
Collapse
|
19
|
Heck MV, Azizov M, Stehning T, Walter M, Kedersha N, Auburger G. Dysregulated expression of lipid storage and membrane dynamics factors in Tia1 knockout mouse nervous tissue. Neurogenetics 2015; 15:135-44. [PMID: 24659297 PMCID: PMC3994287 DOI: 10.1007/s10048-014-0397-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 03/03/2014] [Indexed: 12/13/2022]
Abstract
During cell stress, the transcription and translation of immediate early genes are prioritized, while most other messenger RNAs (mRNAs) are stored away in stress granules or degraded in processing bodies (P-bodies). TIA-1 is an mRNA-binding protein that needs to translocate from the nucleus to seed the formation of stress granules in the cytoplasm. Because other stress granule components such as TDP-43, FUS, ATXN2, SMN, MAPT, HNRNPA2B1, and HNRNPA1 are crucial for the motor neuron diseases amyotrophic lateral sclerosis (ALS)/spinal muscular atrophy (SMA) and for the frontotemporal dementia (FTD), here we studied mouse nervous tissue to identify mRNAs with selective dependence on Tia1 deletion. Transcriptome profiling with oligonucleotide microarrays in comparison of spinal cord and cerebellum, together with independent validation in quantitative reverse transcriptase PCR and immunoblots demonstrated several strong and consistent dysregulations. In agreement with previously reported TIA1 knock down effects, cell cycle and apoptosis regulators were affected markedly with expression changes up to +2-fold, exhibiting increased levels for Cdkn1a, Ccnf, and Tprkb vs. decreased levels for Bid and Inca1 transcripts. Novel and surprisingly strong expression alterations were detected for fat storage and membrane trafficking factors, with prominent +3-fold upregulations of Plin4, Wdfy1, Tbc1d24, and Pnpla2 vs. a −2.4-fold downregulation of Cntn4 transcript, encoding an axonal membrane adhesion factor with established haploinsufficiency. In comparison, subtle effects on the RNA processing machinery included up to 1.2-fold upregulations of Dcp1b and Tial1. The effect on lipid dynamics factors is noteworthy, since also the gene deletion of Tardbp (encoding TDP-43) and Atxn2 led to fat metabolism phenotypes in mouse. In conclusion, genetic ablation of the stress granule nucleator TIA-1 has a novel major effect on mRNAs encoding lipid homeostasis factors in the brain, similar to the fasting effect.
Collapse
Affiliation(s)
- Melanie Vanessa Heck
- Experimental Neurology, Department of Neurology, Goethe University Medical School, Building 89, 3rd floor, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany
| | - Mekhman Azizov
- Experimental Neurology, Department of Neurology, Goethe University Medical School, Building 89, 3rd floor, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany
| | - Tanja Stehning
- Experimental Neurology, Department of Neurology, Goethe University Medical School, Building 89, 3rd floor, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany
| | - Michael Walter
- Institute for Medical Genetics, Eberhard-Karls-University of Tuebingen, 72076 Tübingen, Germany
| | - Nancy Kedersha
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Smith 652, One Jimmy Fund Way, Boston, MA 02115 USA
| | - Georg Auburger
- Experimental Neurology, Department of Neurology, Goethe University Medical School, Building 89, 3rd floor, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
20
|
Mallick S, D'Mello SR. JAZ (Znf346), a SIRT1-interacting protein, protects neurons by stimulating p21 (WAF/CIP1) protein expression. J Biol Chem 2014; 289:35409-20. [PMID: 25331946 DOI: 10.1074/jbc.m114.597575] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SIRT1, a class III histone deacetylase, protects neurons in various models of neurodegenerative diseases. We previously described that neuroprotection by SIRT1 is independent of its catalytic activity. To elucidate how SIRT1 protects neurons, we performed a mass spectrometric screen to find SIRT1-interacting proteins. One of the proteins identified was JAZ (Znf346), a member of a new class of Cys-2-His-2 zinc finger proteins. To investigate the significance of JAZ in the regulation of neuronal survival, we overexpressed it in neurons. We found that JAZ protects cerebellar granule neurons against potassium deprivation-induced death and cortical neurons from death resulting from oxidative stress. JAZ also protects neurons against toxicity induced by mutant huntingtin and mutant ataxin-1 expression. Although expression of endogenous JAZ does not change in neurons primed to die, knockdown of its expression promotes death of otherwise healthy neurons. In contrast to its protective effect in neurons, overexpression of JAZ in different cell lines promotes death. We find that JAZ suppresses cell cycle progression, thereby explaining its contrasting effect in postmitotic neurons versus proliferating cell lines. Although not affecting the expression of several cyclins, overexpression of JAZ stimulates expression of p21 (WAF1/CIP1), a cell cycle inhibitor known to have neuroprotective effects. Results of chromatin immunoprecipitation and transcriptional assays indicate that the stimulatory effect of JAZ on p21 expression is mediated at the transcriptional level. Furthermore, knockdown of p21 expression inhibits the neuroprotective effect of JAZ. Together, our results suggest that JAZ protects neurons by inhibiting cell cycle re-entry through the transcriptional stimulation of p21 expression.
Collapse
Affiliation(s)
- Sathi Mallick
- From the Department of Molecular and Cell Biology, University of Texas at Dallas, Richardson, Texas 75080 and the Department of Biological Sciences, Southern Methodist University, Dallas, Texas 75275
| | - Santosh R D'Mello
- the Department of Biological Sciences, Southern Methodist University, Dallas, Texas 75275
| |
Collapse
|
21
|
Feng Y, Lu S, Wang J, Kumar P, Zhang L, Bhatt AJ. Dexamethasone-induced neuroprotection in hypoxic-ischemic brain injury in newborn rats is partly mediated via Akt activation. Brain Res 2014; 1589:68-77. [PMID: 25304361 DOI: 10.1016/j.brainres.2014.09.073] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 08/28/2014] [Accepted: 09/02/2014] [Indexed: 01/10/2023]
Abstract
Prior treatment with dexamethasone (Dex) provides neuroprotection against hypoxia ischemia (HI) in newborn rats. Recent studies have shown that the phosphatidylinositol-3-kinase/Akt (PI3K/Akt) pathway plays an important role in the neuroprotection. The objective of this study is to evaluate the role of the PI3K/Akt pathway in the Dex-induced neuroprotection against subsequent HI brain injury. Seven-day-old rat pups had the right carotid artery permanently ligated followed by 160min of hypoxia (8% oxygen). Rat pups received i.p. injection of either saline or Dex (0.25mg/kg) at 24 and 4h before HI exposure. To quantify the effects of a PI3K/Akt inhibitor, wortmannin (1μl of 1μg/μl) or vehicle was injected intracerebroventricularly in the right hemisphere on postnatal day 6 at 30min prior to the first dose of Dex or saline treatment. Dex pretreatment significantly reduced the brain injury following HI which was quantified by the decrease in cleaved caspase-3 protein as well as cleaved caspase-3 and TUNEL positive cells at 24h and percent loss of ipsilateral hemisphere weight at 22d after HI, while wortmannin partially reversed these effects. We conclude that Dex provides robust neuroprotection against subsequent HI in newborn rats in part via activation of PI3/Akt pathway.
Collapse
Affiliation(s)
- Yangzheng Feng
- Department of Pediatrics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, USA
| | - Shiqi Lu
- Department of Emergency, The First affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Junming Wang
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Praveen Kumar
- Department of Pediatrics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, USA
| | - Lei Zhang
- Office of Health Data and Research, Mississippi State Department of Health, Jackson, MS 39216, USA
| | - Abhay J Bhatt
- Department of Pediatrics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, USA.
| |
Collapse
|
22
|
Ninomiya E, Hattori T, Toyoda M, Umezawa A, Hamazaki T, Shintaku H. Glucocorticoids promote neural progenitor cell proliferation derived from human induced pluripotent stem cells. SPRINGERPLUS 2014; 3:527. [PMID: 25279318 PMCID: PMC4174547 DOI: 10.1186/2193-1801-3-527] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 09/06/2014] [Indexed: 02/08/2023]
Abstract
Glucocorticoids (GCs) are frequently used for treating and preventing chronic lung disease and circulatory dysfunction in premature infants. However, there is growing concern about the detrimental effects of systemic GC administration on neurodevelopment. The first choice of GCs to minimize the adverse effects on the developing brain is still under debate. We investigated the effect of commonly used GCs such as dexamethasone (DEX), betamethasone (BET) and hydrocortisone (HDC) on the proliferation of human-induced pluripotent stem cell (iPSC)-derived neuronal progenitor cells (NPCs). In this study, NPCs were treated with various concentrations of GCs and subjected to cell proliferation assays. Furthermore, we quantified the number of microtubule-associated protein 2 (MAP2) positive neurons in NPCs by immunostaining. All GCs promoted NPC proliferation in a dose-dependent manner. We also confirmed that MAP2-positive neurons in NPCs increased upon GC treatment. However, differential effects of GCs on MAP2 positive neurons were observed when we treated NPCs with H2O2. The total numbers of NPCs increased upon any GC treatment even under oxidative conditions but the numbers of MAP2 positive neurons increased only by HDC treatment. GCs promoted human iPSCs–derived NPC proliferation and the differential effects of GCs became apparent under oxidative stress. Our results may support HDC as the preferred choice over DEX and BET to prevent adverse effects on the developing human brain.
Collapse
Affiliation(s)
- Eiichi Ninomiya
- Department of Pediatrics, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585 Japan
| | - Taeka Hattori
- Department of Pediatrics, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585 Japan
| | - Masashi Toyoda
- Research Team for Geriatric Medicine (Vascular Medicine), Tokyo Metropolitan Institute of Gerontology, Sakaecho 35-2, Itabashi-Ku, Tokyo, 173-0015 Japan
| | - Akihiro Umezawa
- Department of Reproductive Biology, National Research Institute for Child Health and Development, 2-10-1 Ookura, Setagaya-ku, Tokyo, 157-8535 Japan
| | - Takashi Hamazaki
- Department of Pediatrics, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585 Japan
| | - Haruo Shintaku
- Department of Pediatrics, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585 Japan
| |
Collapse
|
23
|
Chaudhary MW, Al-Baradie RS. Ataxia-telangiectasia: future prospects. APPLICATION OF CLINICAL GENETICS 2014; 7:159-67. [PMID: 25258552 PMCID: PMC4173637 DOI: 10.2147/tacg.s35759] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ataxia-telangiectasia (A-T) is an autosomal recessive multi-system disorder caused by mutation in the ataxia-telangiectasia mutated gene (ATM). ATM is a large serine/threonine protein kinase, a member of the phosphoinositide 3-kinase-related protein kinase (PIKK) family whose best-studied function is as master controller of signal transduction for the DNA damage response (DDR) in the event of double strand breaks (DSBs). The DDR rapidly recognizes DNA lesions and initiates the appropriate cellular programs to maintain genome integrity. This includes the coordination of cell-cycle checkpoints, transcription, translation, DNA repair, metabolism, and cell fate decisions, such as apoptosis or senescence. DSBs can be generated by exposure to ionizing radiation (IR) or various chemical compounds, such as topoisomerase inhibitors, or can be part of programmed generation and repair of DSBs via cellular enzymes needed for the generation of the antibody repertoire as well as the maturation of germ cells. AT patients have immunodeficiency, and are sterile with gonadal dysgenesis as a result of defect in meiotic recombination. In the cells of nervous system ATM has additional role in vesicle dynamics as well as in the maintenance of the epigenetic code of histone modifications. Moderate levels of ATM are associated with prolonged lifespan through resistance to oxidative stress. ATM inhibitors are being viewed as potential radiosensitizers as part of cancer radiotherapy. Though there is no cure for the disease at present, glucocorticoids have been shown to induce alternate splicing site in the gene for ATM partly restoring its activity, but their most effective timing in the disease natural history is not yet known. Gene therapy is promising but large size of the gene makes it technically difficult to be delivered across the blood-brain barrier at present. As of now, apart from glucocorticoids, use of histone deacetylase inhibitors/EZH2 to minimize effect of the absence of ATM, looks more promising.
Collapse
Affiliation(s)
- Mohammed Wajid Chaudhary
- Pediatric Neurology, Neurosciences Centre, King Fahad Specialist Hospital, Dammam, Kingdom of Saudi Arabia
| | - Raidah Saleem Al-Baradie
- Pediatric Neurology, Neurosciences Centre, King Fahad Specialist Hospital, Dammam, Kingdom of Saudi Arabia
| |
Collapse
|
24
|
|
25
|
Forkwa TK, Tamm ER, Ohlmann A. Ambiguous role of glucocorticoids on survival of retinal neurons. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 801:365-71. [PMID: 24664719 DOI: 10.1007/978-1-4614-3209-8_46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Glucocorticoids (GCs) have a wide range of functions on several mammalian cell types, most of which are aimed at boosting survival, which is the raison d'être of the acute stress response. The role GCs play in the survival and viability of neurons is incongruous, as studies have revealed neuroprotective as well as neurodegenerative effects. These effects seem to depend on multiple factors amongst which are; the cell type involved, the mode of injury or underlying cause of cell death, likewise the concentration and or duration of GC exposure.In this mini review, we discuss mechanisms of GC action and their effect on neurodegeneration in general, and specifically review the effect of GCs on retinal neurons, in animal models of retinal degeneration or acute neuronal damage. Finally, we summarize potential protective and harmful GC-mediated mechanisms, which might be involved in the determination of neuronal fate in the retina following injury or during degeneration.
Collapse
Affiliation(s)
- Tembei K Forkwa
- Institute of Human Anatomy and Embryology, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany,
| | | | | |
Collapse
|
26
|
Beinder L, Faehrmann N, Wachtveitl R, Winterfeld I, Hartner A, Menendez-Castro C, Rauh M, Ruebner M, Huebner H, Noegel SC, Doerr HG, Rascher W, Fahlbusch FB. Detection of expressional changes induced by intrauterine growth restriction in the developing rat mammary gland via exploratory pathways analysis. PLoS One 2014; 9:e100504. [PMID: 24955840 PMCID: PMC4067350 DOI: 10.1371/journal.pone.0100504] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 05/26/2014] [Indexed: 12/29/2022] Open
Abstract
Background Intrauterine growth restriction (IUGR) is thought to lead to fetal programming that in turn contributes to developmental changes of many organs postnatally. There is evidence that IUGR is a risk factor for the development of metabolic and cardiovascular disease later in life. A higher incidence of breast cancer was also observed after IUGR. This could be due to changes in mammary gland developmental pathways. We sought to characterise IUGR-induced alterations of the complex pathways of mammary development at the level of the transcriptome in a rat model of IUGR, using pathways analysis bioinformatics. Methodology/Principal Findings We analysed the mammary glands of Wistar rats with IUGR induced by maternal low protein (LP) diet at the beginning (d21) and the end (d28) of pubertal ductal morphogenesis. Mammary glands of the LP group were smaller in size at d28, however did not show morphologic changes. We identified multiple differentially expressed genes in the mammary gland using Agilent SurePrint arrays at d21 and d28. In silico analysis was carried out using Ingenuity Pathways Analysis. In mammary gland tissue of LP rats at d21 of life a prominent upregulation of WT1 and CDKN1A (p21) expression was observed. Differentially regulated genes were associated with the extracellular regulated kinase (ERK)-1/-2 pathway. Western Blot analysis showed reduced levels of phosphorylated ERK-1/-2 in the mammary glands of the LP group at d21. To identify possible changes in circulating steroid levels, serum LC-Tandem mass-spectrometry was performed. LP rats showed higher serum progesterone levels and an increased corticosterone/dehydrocorticosterone-ratio at d28. Conclusions/Significance Our data obtained from gene array analysis support the hypothesis that IUGR influences pubertal development of the rat mammary gland. We identified prominent differential regulation of genes and pathways for factors regulating cell cycle and growth. Moreover, we detected new pathways which appear to be programmed by IUGR.
Collapse
Affiliation(s)
- Lea Beinder
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Nina Faehrmann
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Rainer Wachtveitl
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Ilona Winterfeld
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Andrea Hartner
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Carlos Menendez-Castro
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Ruebner
- Department of Gynecology and Obstetrics, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Hanna Huebner
- Department of Gynecology and Obstetrics, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Stephanie C. Noegel
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Helmuth G. Doerr
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Wolfgang Rascher
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Fabian B. Fahlbusch
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
- * E-mail:
| |
Collapse
|
27
|
Biagiotti S, Menotta M, Giacomini E, Radici L, Bianchi M, Bozzao C, Chessa L, Magnani M. Forward subtractive libraries containing genes transactivated by dexamethasone in ataxia-telangiectasia lymphoblastoid cells. Mol Cell Biochem 2014; 392:13-30. [DOI: 10.1007/s11010-014-2013-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 02/28/2014] [Indexed: 11/30/2022]
|
28
|
Forkwa TK, Neumann ID, Tamm ER, Ohlmann A, Reber SO. Short-term psychosocial stress protects photoreceptors from damage via corticosterone-mediated activation of the AKT pathway. Exp Neurol 2014; 252:28-36. [DOI: 10.1016/j.expneurol.2013.11.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 11/09/2013] [Accepted: 11/14/2013] [Indexed: 01/22/2023]
|
29
|
Abstract
Glucocorticoids (GCs) are frequently prescribed pharmacological agents most notably for their immunosuppressive effects. Endogenous GCs mediate biological processes such as energy metabolism and tissue development. At the cellular level, GCs bind to the glucocorticoid receptor (GR), a cytosolic protein that translocates to the nuclei and functions to alter transcription upon ligand binding. Among a long list of genes activated by GCs is the glucocorticoid-induced leucine zipper (GILZ). GC-induced GILZ expression has been well established in lymphocytes and mediates GC-induced apoptosis. Unlike lymphocytes, cardiomyocytes respond to GCs by gaining resistance against apoptosis. We determined GILZ expression in cardiomyocytes in vivo and in vitro. Expression of GILZ in mouse hearts as a result of GC administration was confirmed by Western blot analyses. GCs induced dose- and time-dependent elevation of GILZ expression in primary cultured rat cardiomyocytes, with dexamethasone (Dex) as low as 0.1 μM being effective. Time course analysis indicated that GILZ protein levels increased at 8 h and peaked at 48 h after exposure to 1 μM Dex. H9c2(2-1) cell line showed a similar response of GILZ induction by Dex as primary cultured rat cardiomyocytes, providing a convenient model for studying the biological significance of GILZ expression. With corticosterone (CT), an endogenous form of corticosteroids in rodents, 0.1-2.5 μM was found to induce GILZ in H9c2(2-1) cells. Time course analysis with 1 μM CT indicated induction of GILZ at 6 h with peak expression at 18 h. Inhibition of the GR by mifepristone led to blunting of GILZ induction by GCs. Our data demonstrate GILZ induction in cardiomyocytes both in vivo and in vitro by GCs, pointing to H9c2(2-1) cells as a valid model for studying the biological function of GILZ in cardiomyocytes.
Collapse
|
30
|
Krolewski RC, Packard A, Schwob JE. Global expression profiling of globose basal cells and neurogenic progression within the olfactory epithelium. J Comp Neurol 2013; 521:833-59. [PMID: 22847514 DOI: 10.1002/cne.23204] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 07/02/2012] [Accepted: 07/25/2012] [Indexed: 01/08/2023]
Abstract
Ongoing, lifelong neurogenesis maintains the neuronal population of the olfactory epithelium in the face of piecemeal neuronal turnover and restores it following wholesale loss. The molecular phenotypes corresponding to different stages along the progression from multipotent globose basal cell (GBC) progenitor to differentiated olfactory sensory neuron are poorly characterized. We used the transgenic expression of enhanced green fluorescent protein (eGFP) and cell surface markers to FACS-isolate ΔSox2-eGFP(+) GBCs, Neurog1-eGFP(+) GBCs and immature neurons, and ΔOMP-eGFP(+) mature neurons from normal adult mice. In addition, the latter two populations were also collected 3 weeks after olfactory bulb ablation, a lesion that results in persistently elevated neurogenesis. Global profiling of mRNA from the populations indicates that all stages of neurogenesis share a cohort of >2,100 genes that are upregulated compared to sustentacular cells. A further cohort of >1,200 genes are specifically upregulated in GBCs as compared to sustentacular cells and differentiated neurons. The increased rate of neurogenesis caused by olfactory bulbectomy had little effect on the transcriptional profile of the Neurog1-eGFP(+) population. In contrast, the abbreviated lifespan of ΔOMP-eGFP(+) neurons born in the absence of the bulb correlated with substantial differences in gene expression as compared to the mature neurons of the normal epithelium. Detailed examination of the specific genes upregulated in the different progenitor populations revealed that the chromatin modifying complex proteins LSD1 and coREST were expressed sequentially in upstream ΔSox2-eGFP(+) GBCs and Neurog1-eGFP(+) GBCs/immature neurons. The expression patterns of these proteins are dynamically regulated after activation of the epithelium by methyl bromide lesion.
Collapse
Affiliation(s)
- Richard C Krolewski
- Department of Anatomy and Cellular Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | |
Collapse
|
31
|
Drakulić D, Veličković N, Stanojlović M, Grković I, Mitrović N, Lavrnja I, Horvat A. Low-dose dexamethasone treatment promotes the pro-survival signalling pathway in the adult rat prefrontal cortex. J Neuroendocrinol 2013; 25:605-16. [PMID: 23551329 DOI: 10.1111/jne.12037] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 02/15/2013] [Accepted: 03/24/2013] [Indexed: 11/29/2022]
Abstract
Synthetic glucocorticoid dexamethasone (DEX), a highly potent anti-inflammatory and immunosuppressive agent, is widely used in the treatment of brain cancer, as well as for inflammatory and autoimmune diseases. The present study aimed to determine whether low-dose subchronic DEX treatment (100 μg/kg for eight consecutive days) exerts long-term effects on apoptosis in the adult rat prefrontal cortex (PFC) by examining the expression of cell death-promoting molecules [poly(ADP-ribose) polymerase (PARP), p53, procaspase 3, cleaved caspase 3, Bax] and cell-survival molecules (AKT, Bcl-2). The results obtained revealed that body, thymus and adrenal gland weights, as well corticosterone levels, in the serum and PFC were reduced 1 day after the last DEX injection. In the PFC, DEX caused activation of AKT, augmentation of pro-survival Bcl-2 protein and an enhanced Bcl-2/Bax protein ratio, as well Bcl-2 translocation to the mitochondria. An unaltered profile with respect to the protein expression of apoptotic molecules PARP, procaspase 3 and Bax was detected, whereas p53 protein was decreased. Reverse transcriptase -polymerase chain reaction analysis showed a decrease of p53 mRNA levels and no significant difference in Bcl-2 and Bax mRNA expression in DEX-treated rats. Finally, a DNA fragmentation assay and Fluoro-Jade staining demonstrated no considerable changes in apoptosis in the rat PFC. Our findings support the concept that low-dose DEX creates a hypocorticoid state in the brain and also indicate that subchronic DEX treatment activates the pro-survival signalling pathway but does not change apoptotic markers in the rat PFC. This mechanism might be relevant for the DEX-induced apoptosis resistance observed during and after chemotherapy of patients with brain tumours.
Collapse
Affiliation(s)
- D Drakulić
- Laboratory of Molecular Biology and Endocrinology, VINCA Institute of Nuclear Sciences, University of Belgrade, 11001 Belgrade, Serbia.
| | | | | | | | | | | | | |
Collapse
|
32
|
Jahn SC, Law ME, Corsino PE, Rowe TC, Davis BJ, Law BK. Assembly, activation, and substrate specificity of cyclin D1/Cdk2 complexes. Biochemistry 2013; 52:3489-501. [PMID: 23627734 DOI: 10.1021/bi400047u] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Previous studies have shown conflicting data regarding cyclin D1/cyclin-dependent kinase 2 (Cdk2) complexes, and considering the widespread overexpression of cyclin D1 in cancer, it is important to fully understand their relevance. While many have shown that cyclin D1 and Cdk2 form active complexes, others have failed to show activity or association. Here, using a novel p21-PCNA fusion protein as well as p21 mutant proteins, we show that p21 is a required scaffolding protein, with cyclin D1 and Cdk2 failing to complex in its absence. These p21/cyclin D1/Cdk2 complexes are active and also bind the trimeric PCNA complex, with each trimer capable of independently binding distinct cyclin/Cdk complexes. We also show that increased p21 levels due to treatment with chemotherapeutic agents result in increased formation and kinase activity of cyclin D1/Cdk2 complexes, and that cyclin D1/Cdk2 complexes are able to phosphorylate a number of substrates in addition to Rb. Nucleophosmin and Cdh1, two proteins important for centrosome replication and implicated in the chromosomal instability of cancer, are shown to be phosphorylated by cyclin D1/Cdk2 complexes. Additionally, polypyrimidine tract binding protein-associated splicing factor (PSF) is identified as a novel Cdk2 substrate, being phosphorylated by Cdk2 complexed with either cyclin E or cyclin D1, and given the many functions of PSF, it could have important implications on cellular activity.
Collapse
Affiliation(s)
- Stephan C Jahn
- Department of Pharmacology and Therapeutics and the ‡Shands Cancer Center, University of Florida , Gainesville, Florida 32610, United States
| | | | | | | | | | | |
Collapse
|
33
|
Kondo MA, Tajinda K, Colantuoni C, Hiyama H, Seshadri S, Huang B, Pou S, Furukori K, Hookway C, Jaaro-Peled H, Kano SI, Matsuoka N, Harada K, Ni K, Pevsner J, Sawa A. Unique pharmacological actions of atypical neuroleptic quetiapine: possible role in cell cycle/fate control. Transl Psychiatry 2013; 3:e243. [PMID: 23549417 PMCID: PMC3641406 DOI: 10.1038/tp.2013.19] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Quetiapine is an atypical neuroleptic with a pharmacological profile distinct from classic neuroleptics that function primarily via blockade of dopamine D2 receptors. In the United States, quetiapine is currently approved for treating patients with schizophrenia, major depression and bipolar I disorder. Despite its widespread use, its cellular effects remain elusive. To address possible mechanisms, we chronically treated mice with quetiapine, haloperidol or vehicle and examined quetiapine-specific gene expression change in the frontal cortex. Through microarray analysis, we observed that several groups of genes were differentially expressed upon exposure to quetiapine compared with haloperidol or vehicle; among them, Cdkn1a, the gene encoding p21, exhibited the greatest fold change relative to haloperidol. The quetiapine-induced downregulation of p21/Cdkn1a was confirmed by real-time polymerase chain reaction and in situ hybridization. Consistent with single gene-level analyses, functional group analyses also indicated that gene sets associated with cell cycle/fate were differentially regulated in the quetiapine-treated group. In cortical cell cultures treated with quetiapine, p21/Cdkn1a was significantly downregulated in oligodendrocyte precursor cells and neurons, but not in astrocytes. We propose that cell cycle-associated intervention by quetiapine in the frontal cortex may underlie a unique efficacy of quetiapine compared with typical neuroleptics.
Collapse
Affiliation(s)
- M A Kondo
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - K Tajinda
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA,Pharmacology Research Labs, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - C Colantuoni
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - H Hiyama
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA,Pharmacology Research Labs, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - S Seshadri
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - B Huang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - S Pou
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - K Furukori
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - C Hookway
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - H Jaaro-Peled
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - S-i Kano
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - N Matsuoka
- Pharmacology Research Labs, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - K Harada
- Pharmacology Research Labs, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - K Ni
- Pharmacology Research Labs, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - J Pevsner
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA,Hugo W Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University and Hugo W Moser Research Institute at Kennedy Krieger, Baltimore, MD 21287, USA. E-mail:
| | - A Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA,Department of Psychiatry, Johns Hopkins University, 600 North Wolfe Street, Meyer 3-166, Baltimore, MD 21287, USA. E-mail:
| |
Collapse
|
34
|
A functional role of the cyclin-dependent kinase inhibitor 1 (p21(WAF1/CIP1)) for neuronal preconditioning. J Cereb Blood Flow Metab 2013; 33:351-5. [PMID: 23299246 PMCID: PMC3587824 DOI: 10.1038/jcbfm.2012.213] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Hypoxic preconditioning is thought to rely on gene products regulated by hypoxia-inducible factor (HIF)-1. Here, we show that the HIF-1 target gene cyclin-dependent kinase inhibitor 1, p21(WAF1/CIP1), is essential for neuroprotection by hypoxic/aglycemic or erythropoietin preconditioning using wild-type and p21(WAF1/CIP1)-deficient neurons. Furthermore, overexpression of wild-type p21(WAF1/CIP1) or phospho-mutants significantly increased cell death after hypoxia/aglycemia. Moreover, deferoxamine-induced endogenous tolerance did not involve p21(WAF1/CIP1) expression in cortical neurons. Our data suggest that balanced expression and potentially posttranslational regulation of p21(WAF1/CIP1) is required for hypoxic preconditioning.
Collapse
|
35
|
Park JH, Lee SB, Lee KH, Ahn JY. Nuclear Akt promotes neurite outgrowth in the early stage of neuritogenesis. BMB Rep 2013; 45:521-5. [PMID: 23010173 DOI: 10.5483/bmbrep.2012.45.9.114] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In addition to its pivotal role in neuronal survival, PI3K/Akt signaling is integral to neuronal differentiation and neurite outgrowth. However, the exact role of Akt in neuronal differentiation is still controversial. Here, we found that nuclear expression of CA-Akt resulted in unusual rapid neurite outgrowth and overexpression of KD-Akt caused multiple dendrite growth without specific axon elongation. Moreover, microarray data revealed that the expression of FOXQ1 expression was about 10-fold higher in cells with nuclear, active Akt than in control cells. Quantitative real-time PCR analysis showed that mRNA levels were upregulated in NLS-CA-Akt cells as compared to KD or EV cells. Furthermore, our FACS analysis demonstrated that overexpression of NLS-CA-Akt accumulate cells in the G1 phase within 24 h, fitting with the rapid sprouting of neuritis. Thus, our data implied that at least in this early time frame, the overexpression of nuclear, active Akt forced cells into neurite development through probably FOXQ1regulation.
Collapse
Affiliation(s)
- Ji Hye Park
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea
| | | | | | | |
Collapse
|
36
|
Koga K, Kenessey A, Ojamaa K. Macrophage migration inhibitory factor antagonizes pressure overload-induced cardiac hypertrophy. Am J Physiol Heart Circ Physiol 2012; 304:H282-93. [PMID: 23144312 DOI: 10.1152/ajpheart.00595.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Macrophage migration inhibitory factor (MIF) functions as a proinflammatory cytokine when secreted from the cell, but it also exhibits antioxidant properties by virtue of its intrinsic oxidoreductase activity. Since increased production of ROS is implicated in the development of left ventricular hypertrophy, we hypothesized that the redox activity of MIF protects the myocardium when exposed to hemodynamic stress. In a mouse model of myocardial hypertrophy induced by transverse aortic coarctation (TAC) for 10 days, we showed that growth of the MIF-deficient heart was significantly greater by 32% compared with wild-type (WT) TAC hearts and that fibrosis was increased by fourfold (2.62 ± 0.2% vs. 0.6 ± 0.1%). Circulating MIF was increased in TAC animals, and expression of MIF receptor, CD74, was increased in the hypertrophic myocardium. Gene expression analysis showed a 10-fold increase (P < 0.01) in ROS-generating mitochondrial NADPH oxidase and 2- to 3-fold reductions (P < 0.01) in mitochondrial SOD2 and mitochondrial aconitase activities, indicating enhanced oxidative injury in the hypertrophied MIF-deficient ventricle. Hypertrophic signaling pathways showed that phosphorylation of cytosolic glycogen synthase kinase-3α was greater (P < 0.05) at baseline in MIF-deficient hearts than in WT hearts and remained elevated after 10-day TAC. In the hemodynamically stressed MIF-deficient heart, nuclear p21(CIP1) increased sevenfold (P < 0.01), and the cytosolic increase of phospho-p21(CIP1) was significantly greater than in WT TAC hearts. We conclude that MIF antagonizes myocardial hypertrophy and fibrosis in response to hemodynamic stress by maintaining a redox homeostatic phenotype and attenuating stress-induced activation of hypertrophic signaling pathways.
Collapse
Affiliation(s)
- Kiyokazu Koga
- Center for Heart and Lung Research, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | | | | |
Collapse
|
37
|
Giardino G, Fusco A, Romano R, Gallo V, Maio F, Esposito T, Palamaro L, Parenti G, Salerno MC, Vajro P, Pignata C. Betamethasone therapy in ataxia telangiectasia: unraveling the rationale of this serendipitous observation on the basis of the pathogenesis. Eur J Neurol 2012; 20:740-7. [PMID: 23121321 DOI: 10.1111/ene.12024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 09/20/2012] [Indexed: 11/30/2022]
Abstract
Ataxia telangiectasia (A-T) is a rare autosomal recessive disorder characterized by progressive neurological dysfunction. To date, only supportive care aimed to halt the progressive neurodegeneration is available for the treatment. Recently, an improvement of neurological signs during short-term treatment with betamethasone has been reported. To date, the molecular and biochemical mechanisms by which the steroid produces such effects have not yet been elucidated. Therefore, a review of the literature was carried out to define the potential molecular and functional targets of the steroid effects in A-T. Glucocorticoids (GCs) are capable of diffusing into the CNS by crossing the blood-brain barrier (BBB) where they exert effects on the suppression of inflammation or as antioxidant. GCs have been shown to protect post-mitotic neurons from apoptosis. Eventually, GCs may also modulate synaptic plasticity. A better understanding of the mechanisms of action of GCs in the brain is needed, because in A-T during the initial phase of cell loss the neurological impairment may be rescued by interfering in the biochemical pathways. This would open a new window of intervention in this so far incurable disease.
Collapse
Affiliation(s)
- G Giardino
- Department of Pediatrics, Federico II University, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Ma TC, Langley B, Ko B, Wei N, Gazaryan IG, Zareen N, Yamashiro DJ, Willis DE, Ratan RR. A screen for inducers of p21(waf1/cip1) identifies HIF prolyl hydroxylase inhibitors as neuroprotective agents with antitumor properties. Neurobiol Dis 2012; 49:13-21. [PMID: 22944173 DOI: 10.1016/j.nbd.2012.08.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 08/08/2012] [Accepted: 08/16/2012] [Indexed: 11/15/2022] Open
Abstract
Preventing neuronal death is a priority for treating neurological diseases. However, therapies that inhibit pathological neuron loss could promote tumorigenesis by preventing the physiological death of cancerous cells. To avert this, we targeted the transcriptional upregulation of p21(waf1/cip1) (p21), an endogenous tumor suppressor with neuroprotective and pro-regenerative activity. We identified potential p21 indcuers by screening a FDA-approved drug and natural product small molecule library against hippocampal HT22 cells stably expressing a luciferase reporter driven by the proximal 60bp of the p21 promoter, and tested them for neuroprotection from glutathione depletion mediated oxidative stress, and cytotoxicity to cancer cell lines (DLD-1, Neuro-2A, SH-SY5Y, NGP, CHLA15, CHP212, and SK-N-SH) in vitro. Of the p21 inducers identified, only ciclopirox, a hypoxia-inducible factor prolyl-4-hydroxylase (HIF-PHD) inhibitor, simultaneously protected neurons from glutathione depletion and decreased cancer cell proliferation at concentrations that were not basally toxic to neurons. We found that other structurally distinct HIF-PHD inhibitors (desferrioxamine, 3,4-dihydroxybenzoate, and dimethyloxalyl glycine) also protected neurons at concentrations that killed cancer cells. HIF-PHD inhibitors stabilize HIF transcription factors, mediating genetic adaptation to hypoxia. While augmenting HIF stability is believed to promote tumorigenesis, we found that chronic HIF-PHD inhibition killed cancer cells, suggesting a protumorigenic role for these enzymes. Moreover, our findings suggest that PHD inhibitors can be used to treat neurological disease without significant concern for cell-autonomous tumor promotion.
Collapse
Affiliation(s)
- Thong C Ma
- Burke-Cornell Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, USA; Department of Neurology and Neuroscience, Weill Medical College of Cornell University, 525 East 68th Street, New York, NY 10065, USA.
| | - Brett Langley
- Burke-Cornell Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, USA; Department of Neurology and Neuroscience, Weill Medical College of Cornell University, 525 East 68th Street, New York, NY 10065, USA
| | - Brian Ko
- Burke-Cornell Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, USA; Department of Neurology and Neuroscience, Weill Medical College of Cornell University, 525 East 68th Street, New York, NY 10065, USA
| | - Na Wei
- Department of Pediatrics, Pathology, and Cell Biology, Columbia University College of Physicians and Surgeons, 161 Fort Washington Avenue, New York, NY 10032, USA
| | - Irina G Gazaryan
- Burke-Cornell Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, USA; Department of Neurology and Neuroscience, Weill Medical College of Cornell University, 525 East 68th Street, New York, NY 10065, USA
| | - Neela Zareen
- Department of Pathology, Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY, 10032, USA
| | - Darrell J Yamashiro
- Department of Pediatrics, Pathology, and Cell Biology, Columbia University College of Physicians and Surgeons, 161 Fort Washington Avenue, New York, NY 10032, USA
| | - Dianna E Willis
- Burke-Cornell Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, USA; Department of Neurology and Neuroscience, Weill Medical College of Cornell University, 525 East 68th Street, New York, NY 10065, USA
| | - Rajiv R Ratan
- Burke-Cornell Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, USA; Department of Neurology and Neuroscience, Weill Medical College of Cornell University, 525 East 68th Street, New York, NY 10065, USA.
| |
Collapse
|
39
|
Gertz K, Kronenberg G, Kälin RE, Baldinger T, Werner C, Balkaya M, Eom GD, Hellmann-Regen J, Kröber J, Miller KR, Lindauer U, Laufs U, Dirnagl U, Heppner FL, Endres M. Essential role of interleukin-6 in post-stroke angiogenesis. ACTA ACUST UNITED AC 2012; 135:1964-80. [PMID: 22492561 DOI: 10.1093/brain/aws075] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Ambivalent effects of interleukin-6 on the pathogenesis of ischaemic stroke have been reported. However, to date, the long-term actions of interleukin-6 after stroke have not been investigated. Here, we subjected interleukin-6 knockout (IL-6(-/-)) and wild-type control mice to mild brain ischaemia by 30-min filamentous middle cerebral artery occlusion/reperfusion. While ischaemic tissue damage was comparable at early time points, IL-6(-/-) mice showed significantly increased chronic lesion volumes as well as worse long-term functional outcome. In particular, IL-6(-/-) mice displayed an impaired angiogenic response to brain ischaemia with reduced numbers of newly generated endothelial cells and decreased density of perfused microvessels along with lower absolute regional cerebral blood flow and reduced vessel responsivity in ischaemic striatum at 4 weeks. Similarly, the early genomic activation of angiogenesis-related gene networks was strongly reduced and the ischaemia-induced signal transducer and activator of transcription 3 activation observed in wild-type mice was almost absent in IL-6(-/-) mice. In addition, systemic neoangiogenesis was impaired in IL-6(-/-) mice. Transplantation of interleukin-6 competent bone marrow into IL-6(-/-) mice (IL-6(chi)) did not rescue interleukin-6 messenger RNA expression or the early transcriptional activation of angiogenesis after stroke. Accordingly, chronic stroke outcome in IL-6(chi) mice recapitulated the major effects of interleukin-6 deficiency on post-stroke regeneration with significantly enhanced lesion volumes and reduced vessel densities. Additional in vitro experiments yielded complementary evidence, which showed that after stroke resident brain cells serve as the major source of interleukin-6 in a self-amplifying network. Treatment of primary cortical neurons, mixed glial cultures or immortalized brain endothelia with interleukin 6-induced robust interleukin-6 messenger RNA transcription in each case, whereas oxygen-glucose deprivation did not. However, oxygen-glucose deprivation of organotypic brain slices resulted in strong upregulation of interleukin-6 messenger RNA along with increased transcription of key angiogenesis-associated genes. In conclusion, interleukin-6 produced locally by resident brain cells promotes post-stroke angiogenesis and thereby affords long-term histological and functional protection.
Collapse
Affiliation(s)
- Karen Gertz
- Department of Neurology, Charité– Universitätsmedizin Berlin, 10117 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Turban S, Liu X, Ramage L, Webster SP, Walker BR, Dunbar DR, Mullins JJ, Seckl JR, Morton NM. Optimal elevation of β-cell 11β-hydroxysteroid dehydrogenase type 1 is a compensatory mechanism that prevents high-fat diet-induced β-cell failure. Diabetes 2012; 61:642-52. [PMID: 22315313 PMCID: PMC3282808 DOI: 10.2337/db11-1054] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Type 2 diabetes ultimately results from pancreatic β-cell failure. Abnormally elevated intracellular regeneration of glucocorticoids by the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) in fat or liver may underlie pathophysiological aspects of the metabolic syndrome. Elevated 11β-HSD1 is also found in pancreatic islets of obese/diabetic rodents and is hypothesized to suppress insulin secretion and promote diabetes. To define the direct impact of elevated pancreatic β-cell 11β-HSD1 on insulin secretion, we generated β-cell-specific, 11β-HSD1-overexpressing (MIP-HSD1) mice on a strain background prone to β-cell failure. Unexpectedly, MIP-HSD1(tg/+) mice exhibited a reversal of high fat-induced β-cell failure through augmentation of the number and intrinsic function of small islets in association with induction of heat shock, protein kinase A, and extracellular signal-related kinase and p21 signaling pathways. 11β-HSD1(-/-) mice showed mild β-cell impairment that was offset by improved glucose tolerance. The benefit of higher β-cell 11β-HSD1 exhibited a threshold because homozygous MIP-HSD1(tg/tg) mice and diabetic Lep(db/db) mice with markedly elevated β-cell 11β-HSD1 levels had impaired basal β-cell function. Optimal elevation of β-cell 11β-HSD1 represents a novel biological mechanism supporting compensatory insulin hypersecretion rather than exacerbating metabolic disease. These findings have immediate significance for current therapeutic strategies for type 2 diabetes.
Collapse
Affiliation(s)
- Sophie Turban
- Molecular Metabolism Group, University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Xiaoxia Liu
- Molecular Metabolism Group, University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Lynne Ramage
- Molecular Metabolism Group, University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Scott P. Webster
- Endocrinology Unit, University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Brian R. Walker
- Endocrinology Unit, University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Donald R. Dunbar
- Bioinformatics Core, University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - John J. Mullins
- Molecular Physiology, University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Jonathan R. Seckl
- Endocrinology Unit, University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Nicholas M. Morton
- Molecular Metabolism Group, University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, U.K
- Corresponding author: Nicholas M. Morton,
| |
Collapse
|
41
|
Datwyler AL, Lättig-Tünnemann G, Yang W, Paschen W, Lee SLL, Dirnagl U, Endres M, Harms C. SUMO2/3 conjugation is an endogenous neuroprotective mechanism. J Cereb Blood Flow Metab 2011; 31:2152-9. [PMID: 21863037 PMCID: PMC3210338 DOI: 10.1038/jcbfm.2011.112] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Small ubiquitin-like modifier (SUMO)2/3 but not SUMO1 conjugation is activated after transient cerebral ischemia. To investigate its function, we blocked neuronal SUMO2/3 translation through lentiviral microRNA delivery in primary cortical neurons. Viability was unaffected by SUMO2/3 silencing unless neurons were stressed by transient oxygen-glucose deprivation (OGD). Both 15 and 45 minutes of OGD were tolerated by control microRNA-expressing neurons but damaged >60% of neurons expressing SUMO2/3 microRNA. Damaging OGD (75 minutes) increased neuronal loss to 54% (control microRNA) and to 99% (SUMO2/3 microRNA). This suggests that activation of SUMO2/3 conjugation is an endogenous neuroprotective stress response.
Collapse
Affiliation(s)
- Anna Lena Datwyler
- Center for Stroke Research Berlin, Department of Experimental Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Xu H, Lu A, Sharp FR. Regional genome transcriptional response of adult mouse brain to hypoxia. BMC Genomics 2011; 12:499. [PMID: 21988864 PMCID: PMC3218040 DOI: 10.1186/1471-2164-12-499] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Accepted: 10/11/2011] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Since normal brain function depends upon continuous oxygen delivery and short periods of hypoxia can precondition the brain against subsequent ischemia, this study examined the effects of brief hypoxia on the whole genome transcriptional response in adult mouse brain. RESULT Pronounced changes of gene expression occurred after 3 hours of hypoxia (8% O(2)) and after 1 hour of re-oxygenation in all brain regions. The hypoxia-responsive genes were predominantly up-regulated in hindbrain and predominantly down-regulated in forebrain - possibly to support hindbrain survival functions at the expense of forebrain cognitive functions. The up-regulated genes had a significant role in cell survival and involved both shared and unshared signaling pathways among different brain regions. Up-regulation of transcriptional signaling including hypoxia inducible factor, insulin growth factor (IGF), the vitamin D3 receptor/retinoid X nuclear receptor, and glucocorticoid signaling was common to many brain regions. However, many of the hypoxia-regulated target genes were specific for one or a few brain regions. Cerebellum, for example, had 1241 transcripts regulated by hypoxia only in cerebellum but not in hippocampus; and, 642 (54%) had at least one hepatic nuclear receptor 4A (HNF4A) binding site and 381 had at least two HNF4A binding sites in their promoters. The data point to HNF4A as a major hypoxia-responsive transcription factor in cerebellum in addition to its known role in regulating erythropoietin transcription. The genes unique to hindbrain may play critical roles in survival during hypoxia. CONCLUSION Differences of forebrain and hindbrain hypoxia-responsive genes may relate to suppression of forebrain cognitive functions and activation of hindbrain survival functions, which may coordinately mediate the neuroprotection afforded by hypoxia preconditioning.
Collapse
Affiliation(s)
- Huichun Xu
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Bethesda, MD 20892-5635, USA.
| | | | | |
Collapse
|
43
|
Moon DO, Choi YH, Kim GY. Role of p21 in SP600125-induced cell cycle arrest, endoreduplication, and apoptosis. Cell Mol Life Sci 2011; 68:3249-60. [PMID: 21311948 PMCID: PMC11114892 DOI: 10.1007/s00018-011-0626-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Revised: 12/24/2010] [Accepted: 01/06/2011] [Indexed: 11/28/2022]
Abstract
The anti-cancer effect of the c-Jun N-terminal kinase (JNK) inhibitor SP600125 has been well evaluated in human cancer cells. However the role of p21 in SP600125-mediated G(2)/M distribution is not fully understood. Our results showed that the transcriptional activation of p21 by SP600125 is mediated through the proximal regions of multiple Sp1 sites in the p21 promoter following ERK-dependent phosphorylation of Sp1. In this process, p21 induces endoreduplication through the inhibition of cyclin E/Cdk2 activity at 24 h but does not directly regulate cyclin B1/Cdc2 activity. Furthermore, SP600125 induces the phosphorylation of p21 at Thr 145 through the PI3K/Akt pathway. Akt-mediated phosphorylation of p21 and protection of apoptosis are completely abolished by inhibitors of PI3K and Akt. In summary using time points, we identified the dual functions of p21 as an inhibitor of cell-cycle progression at 24 h and as an anti-apoptotic factor at 48 h.
Collapse
Affiliation(s)
- Dong-Oh Moon
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju, 690-756 Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dongeui University, Busan, 614-054 Republic of Korea
| | - Gi-Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju, 690-756 Republic of Korea
| |
Collapse
|
44
|
Berg DA, Kirkham M, Wang H, Frisén J, Simon A. Dopamine controls neurogenesis in the adult salamander midbrain in homeostasis and during regeneration of dopamine neurons. Cell Stem Cell 2011; 8:426-33. [PMID: 21474106 DOI: 10.1016/j.stem.2011.02.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 12/10/2010] [Accepted: 01/14/2011] [Indexed: 11/19/2022]
Abstract
Appropriate termination of regenerative processes is critical for producing the correct number of cells in tissues. Here we provide evidence for an end-product inhibition of dopamine neuron regeneration that is mediated by dopamine. Ablation of midbrain dopamine neurons leads to complete regeneration in salamanders. Regeneration involves extensive neurogenesis and requires activation of quiescent ependymoglia cells, which express dopamine receptors. Pharmacological compensation for dopamine loss by L-dopa inhibits ependymoglia proliferation and regeneration in a dopamine receptor-signaling-dependent manner, specifically after ablation of dopamine neurons. Systemic administration of the dopamine receptor antagonist haloperidol alone causes ependymoglia proliferation and the appearance of excessive number of neurons. Our data show that stem cell quiescence is under dopamine control and provide a model for termination once normal homeostasis is restored. The findings establish a role for dopamine in the reversible suppression of neurogenesis in the midbrain and have implications for regenerative strategies in Parkinson's disease.
Collapse
Affiliation(s)
- Daniel A Berg
- Department of Cell and Molecular Biology and Center of Developmental Biology for Regenerative Medicine (DBRM), Karolinska Institute, SE-171 77 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
45
|
Macrophage migration inhibitory factor promotes cell death and aggravates neurologic deficits after experimental stroke. J Cereb Blood Flow Metab 2011; 31:1093-106. [PMID: 21063426 PMCID: PMC3070968 DOI: 10.1038/jcbfm.2010.194] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Multiple mechanisms contribute to tissue demise and functional recovery after stroke. We studied the involvement of macrophage migration inhibitory factor (MIF) in cell death and development of neurologic deficits after experimental stroke. Macrophage migration inhibitory factor is upregulated in the brain after cerebral ischemia, and disruption of the Mif gene in mice leads to a smaller infarct volume and better sensory-motor function after transient middle cerebral artery occlusion (tMCAo). In mice subjected to tMCAo, we found that MIF accumulates in neurons of the peri-infarct region, particularly in cortical parvalbumin-positive interneurons. Likewise, in cultured cortical neurons exposed to oxygen and glucose deprivation, MIF levels increase, and inhibition of MIF by (S,R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid methyl ester (ISO-1) protects against cell death. Deletion of MIF in Mif(-/-) mice does not affect interleukin-1β protein levels in the brain and serum after tMCAo. Furthermore, disruption of the Mif gene in mice does not affect CD68, but it is associated with higher galectin-3 immunoreactivity in the brain after tMCAo, suggesting that MIF affects the molecular/cellular composition of the macrophages/microglia response after experimental stroke. We conclude that MIF promotes neuronal death and aggravates neurologic deficits after experimental stroke, which implicates MIF in the pathogenesis of neuronal injury after stroke.
Collapse
|
46
|
Abstract
Death receptor (DR) signaling has a major impact on the outcome of numerous neurological diseases, including ischemic stroke. DRs mediate not only cell death signals, but also proinflammatory responses and cell proliferation. Identification of regulatory proteins that control the switch between apoptotic and alternative DR signaling opens new therapeutic opportunities. Fas apoptotic inhibitory molecule 2 (Faim2) is an evolutionary conserved, neuron-specific inhibitor of Fas/CD95-mediated apoptosis. To investigate its role during development and in disease models, we generated Faim2-deficient mice. The ubiquitous null mutation displayed a viable and fertile phenotype without overt deficiencies. However, lack of Faim2 caused an increase in susceptibility to combined oxygen-glucose deprivation in primary neurons in vitro as well as in caspase-associated cell death, stroke volume, and neurological impairment after cerebral ischemia in vivo. These processes were rescued by lentiviral Faim2 gene transfer. In summary, we provide evidence that Faim2 is a novel neuroprotective molecule in the context of cerebral ischemia.
Collapse
|
47
|
Dexamethasone pre-treatment protects brain against hypoxic-ischemic injury partially through up-regulation of vascular endothelial growth factor A in neonatal rats. Neuroscience 2011; 179:223-32. [PMID: 21277350 DOI: 10.1016/j.neuroscience.2011.01.050] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 01/21/2011] [Accepted: 01/22/2011] [Indexed: 10/18/2022]
Abstract
Dexamethasone (Dex) provides neuroprotection against subsequent hypoxia ischemia (HI) in newborn rats, but the mechanism of this neuroprotection is not well understood. It is known that vascular endothelial growth factor A (VEGF) has neuroprotective effects. The objective of this study was to evaluate the role of the VEGF signaling pathway in the Dex-induced neuroprotection in newborn rats. Seven-day-old rat pups had the right carotid artery permanently ligated followed by 140 or 160 min of hypoxia (8% oxygen). Rat pups received two i.p. injections of either saline or Dex (0.25 mg/kg) at 24 and 4 h before HI exposure. To quantify the effects of a glucocorticoid receptor (GR) blocker, on postnatal day (PD) 6 and 15 min prior to Dex treatment rat pups received s.c. vehicle or RU486 (GR blocker, 60 mg/kg). After 24 h at PD 7, all rat pups had HI as described earlier. To quantify the effects of a VEGFR 2 blocker, at 24 h after Dex/Veh treatment (PD7), SU5416, a VEGFR 2 inhibitor or vehicle was injected intracerebroventricularly in the right hemisphere at 30 min before and 2 h after HI. Dex pre-treatment reduced brain injury and enhanced the HI-induced brain VEGF protein while a GR blocker inhibited these effects. Treatment with VEGFR 2 blocker decreased Dex-induced neuroprotection also. Dex pre-treatment enhanced the HI-induced increase in mRNA expression of VEGF splice variants and decreased the HI-induced reduction of Akt phosphorylation. Additionally, it also decreased HI-induced increase of caspase-3 activity and DNA fragments in neonatal rat brain. We conclude that Dex provides robust neuroprotection against subsequent HI in newborn rats via GR likely with the partial involvement of VEGF signaling pathway.
Collapse
|
48
|
Poplawski MM, Mastaitis JW, Mobbs CV. Naloxone, but not valsartan, preserves responses to hypoglycemia after antecedent hypoglycemia: role of metabolic reprogramming in counterregulatory failure. Diabetes 2011; 60:39-46. [PMID: 20811039 PMCID: PMC3012195 DOI: 10.2337/db10-0326] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Hypoglycemia-associated autonomic failure (HAAF) constitutes one of the main clinical obstacles to optimum treatment of type 1 diabetes. Neurons in the ventromedial hypothalamus are thought to mediate counterregulatory responses to hypoglycemia. We have previously hypothesized that hypoglycemia-induced hypothalamic angiotensin might contribute to HAAF, suggesting that the angiotensin blocker valsartan might prevent HAAF. On the other hand, clinical studies have demonstrated that the opioid receptor blocker naloxone ameliorates HAAF. The goal of this study was to generate novel hypothalamic markers of hypoglycemia and use them to assess mechanisms mediating HAAF and its reversal. RESEARCH DESIGN AND METHODS Quantitative PCR was used to validate a novel panel of hypothalamic genes regulated by hypoglycemia. Mice were exposed to one or five episodes of insulin-induced hypoglycemia, with or without concurrent exposure to valsartan or naloxone. Corticosterone, glucagon, epinephrine, and hypothalamic gene expression were assessed after the final episode of hypoglycemia. RESULTS A subset of hypothalamic genes regulated acutely by hypoglycemia failed to respond after repetitive hypoglycemia. Responsiveness of a subset of these genes was preserved by naloxone but not valsartan. Notably, hypothalamic expression of four genes, including pyruvate dehydrogenase kinase 4 and glycerol 3-phosphate dehydrogenase 1, was acutely induced by a single episode of hypoglycemia, but not after antecedent hypoglycemia; naloxone treatment prevented this failure. Similarly, carnitine palmitoyltransferase-1 was inhibited after repetitive hypoglycemia, and this inhibition was prevented by naloxone. Repetitive hypoglycemia also caused a loss of hypoglycemia-induced elevation of glucocorticoid secretion, a failure prevented by naloxone but not valsartan. CONCLUSIONS Based on these observations we speculate that acute hypoglycemia induces reprogramming of hypothalamic metabolism away from glycolysis toward β-oxidation, HAAF is associated with a reversal of this reprogramming, and naloxone preserves some responses to hypoglycemia by preventing this reversal.
Collapse
Affiliation(s)
- Michal M. Poplawski
- Fishberg Center for Neurobiology, Mount Sinai School of Medicine, New York, New York
| | - Jason W. Mastaitis
- Department of Internal Medicine, The Anlyan Center, Yale University School of Medicine, New Haven, Connecticut
| | - Charles V. Mobbs
- Fishberg Center for Neurobiology, Mount Sinai School of Medicine, New York, New York
- Corresponding author: Charles V. Mobbs,
| |
Collapse
|
49
|
Kfir-Erenfeld S, Sionov RV, Spokoini R, Cohen O, Yefenof E. Protein kinase networks regulating glucocorticoid-induced apoptosis of hematopoietic cancer cells: fundamental aspects and practical considerations. Leuk Lymphoma 2010; 51:1968-2005. [PMID: 20849387 DOI: 10.3109/10428194.2010.506570] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glucocorticoids (GCs) are integral components in the treatment protocols of acute lymphoblastic leukemia, multiple myeloma, and non-Hodgkin lymphoma owing to their ability to induce apoptosis of these malignant cells. Resistance to GC therapy is associated with poor prognosis. Although they have been used in clinics for decades, the signal transduction pathways involved in GC-induced apoptosis have only partly been resolved. Accumulating evidence shows that this cell death process is mediated by a communication between nuclear GR affecting gene transcription of pro-apoptotic genes such as Bim, mitochondrial GR affecting the physiology of the mitochondria, and the protein kinase glycogen synthase kinase-3 (GSK3), which interacts with Bim following exposure to GCs. Prevention of Bim up-regulation, mitochondrial GR translocation, and/or GSK3 activation are common causes leading to GC therapy failure. Various protein kinases positively regulating the pro-survival Src-PI3K-Akt-mTOR and Raf-Ras-MEK-ERK signal cascades have been shown to be activated in malignant leukemic cells and antagonize GC-induced apoptosis by inhibiting GSK3 activation and Bim expression. Targeting these protein kinases has proven effective in sensitizing GR-positive malignant lymphoid cells to GC-induced apoptosis. Thus, intervening with the pro-survival kinase network in GC-resistant cells should be a good means of improving GC therapy of hematopoietic malignancies.
Collapse
Affiliation(s)
- Shlomit Kfir-Erenfeld
- The Lautenberg Center of Immunology and Cancer Research, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | | | | | | | |
Collapse
|
50
|
Koster R, di Pietro A, Timmer-Bosscha H, Gibcus JH, van den Berg A, Suurmeijer AJ, Bischoff R, Gietema JA, de Jong S. Cytoplasmic p21 expression levels determine cisplatin resistance in human testicular cancer. J Clin Invest 2010; 120:3594-605. [PMID: 20811155 DOI: 10.1172/jci41939] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Accepted: 07/14/2010] [Indexed: 12/28/2022] Open
Abstract
Platinum-based chemotherapies such as cisplatin are used as first-line treatment for many cancers. Although there is often a high initial responsiveness, the majority of patients eventually relapse with platinum-resistant disease. For example, a subset of testicular cancer patients still die even though testicular cancer is considered a paradigm of cisplatin-sensitive solid tumors, but the mechanisms of chemoresistance remain elusive. Here, we have shown that one key determinant of cisplatin-resistance in testicular embryonal carcinoma (EC) is high cytoplasmic expression of the cyclin-dependent kinase (CDK) inhibitor p21. The EC component of the majority of refractory testicular cancer patients exhibited high cytoplasmic p21 expression, which protected EC cell lines against cisplatin-induced apoptosis via CDK2 inhibition. Localization of p21 in the cytoplasm was critical for cisplatin resistance, since relocalization of p21 to the nucleus by Akt inhibition sensitized EC cell lines to cisplatin. We also demonstrated in EC cell lines and human tumor tissue that high cytoplasmic p21 expression and cisplatin resistance of EC were inversely associated with the expression of Oct4 and miR-106b seed family members. Thus, targeting cytoplasmic p21, including by modulation of the Oct4/miR-106b/p21 pathway, may offer new strategies for the treatment of chemoresistant testicular and other types of cancer.
Collapse
Affiliation(s)
- Roelof Koster
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|