1
|
Lee HS, Lee J, An HJ, Sung MJ, Heo JH, Lee SY, Song YS. Mitophagy Defects Exacerbate Inflammation and Aberrant Proliferation in Lymphocytic Thyroiditis. Thyroid 2024; 34:1401-1413. [PMID: 39397581 DOI: 10.1089/thy.2024.0125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Background: Mitochondrial dysfunction in the thyroid due to defective mitophagy has been observed in lymphocytic thyroiditis (LT). However, the effect of impaired mitophagy on the pathogenesis of LT is not well understood. The aim of this study is to investigate the role of mitophagy dysregulation in the thyroid gland. Methods: We analyzed RNA sequencing data of human thyroid glands with/without LT from Genotype-Tissue Expression (GTEx; n = 653) and performed RNA sequencing in thyroid glands of phosphatase and tensin homolog-induced putative protein kinase 1 (Pink1) knock-out and wild-type mice. We evaluated the phenotypic and histopathologic characteristics of the human (n = 16) and mouse thyroids. Additionally, we assessed cell proliferation, reactive oxygen species (ROS) production, and cytokine secretion of human thyroid epithelial cells (HTori-3) treated with PINK1 siRNA or a mitophagy inhibitor. Results: We found that expression of PINK1, a key regulator of mitophagy, was compromised in human thyroids with LT. Thyroid glands of Pink1-deficient mice exhibited increased inflammatory responses and nodular hyperplasia. Furthermore, mitophagy defects led to the production of pro-inflammatory cytokines and ROS in thyroid cells, resulting in immune cell recruitment. Notably, these mitophagy defects upregulated both the RNA expression and protein secretion of amphiregulin (AREG), an epidermal growth factor receptor (EGFR) ligand, in thyroid cells, while decreasing the protein expression of cAMP response element-binding protein (CREB), a transcription factor that suppresses AREG transcription. Finally, we demonstrated that aberrant cell proliferation in thyroid cells, driven by mitophagy defects, was mitigated after treatment with cetuximab, an EGFR inhibitor. Conclusions: In this study, we observed that mitophagy defects in the thyroid not only intensify inflammation through the accumulation of ROS, cytokine production, and immune cell recruitment but also contribute to hyperplasia via the EGFR pathway, facilitated by increased secretion of AREG from thyroid cells.
Collapse
Affiliation(s)
- Han Sai Lee
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Science, Graduate School, CHA University, Seongnam, South Korea
| | - Jinju Lee
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Science, Graduate School, CHA University, Seongnam, South Korea
| | - Hyun-Ju An
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Min-Ji Sung
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jin-Hyung Heo
- Department of Pathology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - So-Young Lee
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Young Shin Song
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Science, Graduate School, CHA University, Seongnam, South Korea
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| |
Collapse
|
2
|
Barata P, Camacho O, Lima CG, Pereira AC. The Role of Hyperbaric Oxygen Therapy in Neuroregeneration and Neuroprotection: A Review. Cureus 2024; 16:e62067. [PMID: 38989389 PMCID: PMC11235151 DOI: 10.7759/cureus.62067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Neurogenesis is a high energy-demanding process, which is why blood vessels are an active part of the neurogenic niche since they allow the much-needed oxygenation of progenitor cells. In this regard, although neglected for a long time, the "oxygen niche" should be considered an important intervenient in adult neurogenesis. One possible hypothesis for the failure of numerous neuroprotective trials is that they relied on compounds that target a highly specific neuroprotective pathway. This approach may be too limited, given the complexity of the processes that lead to cell death. Therefore, research should adopt a more multifactorial approach. Among the limited range of agents with multimodal neuromodulatory capabilities, hyperbaric oxygen therapy has demonstrated effectiveness in reducing secondary brain damage in various brain injury models. This therapy functions not only as a neuroprotective mechanism but also as a powerful neuroregenerative mechanism.
Collapse
Affiliation(s)
- Pedro Barata
- Pathology and Laboratory Medicine, Centro Hospitalar Universitário do Porto, Porto, PRT
- CECLIN (Center for Clinical Studies), Hospital-Escola da Universidade Fernando Pessoa (HE-UFP), Porto, PRT
| | - Oscar Camacho
- Hyperbaric Medicine Unit, Unidade Local de Saúde de Matosinhos, Matosinhos, PRT
| | - Clara G Lima
- Anesthesiology, Hospital Pedro Hispano, Matosinhos, PRT
| | - Ana Claudia Pereira
- Faculty of Health Sciences, Universidade Fernando Pessoa (UFP), Porto, PRT
- CECLIN (Center for Clinical Studies), Hospital-Escola da Universidade Fernando Pessoa (HE-UFP), Porto, PRT
| |
Collapse
|
3
|
Azzam HN, El-Derany MO, Wahdan SA, Faheim RM, Helal GK, El-Demerdash E. The role of mitochondrial/metabolic axis in development of tamoxifen resistance in breast cancer. Hum Cell 2023; 36:1877-1886. [PMID: 37646973 PMCID: PMC10587280 DOI: 10.1007/s13577-023-00977-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 08/20/2023] [Indexed: 09/01/2023]
Abstract
Only a few investigations, to our knowledge, have examined the bioenergetics of Tamoxifen (TMX) resistant individuals and reported altered mitochondrial activity and metabolic profile. The primary cause of TMX resistance is firmly suggested to be metabolic changes. Metabolic variations and hypoxia have also been linked in a bidirectional manner. Increased hypoxic levels correlate with early recurrence and proliferation and have a negative therapeutic impact on breast cancer (BC) patients. Hypoxia, carcinogenesis, and patient death are all correlated, resulting in more aggressive traits, a higher chance of metastasis, and TMX resistance. Consequently, we sought to investigate the possible role of the metabolic/hypoxial axis Long non-coding RNA (LncRNA) Taurine up-regulated 1 (TUG-1), Micro-RNA 186-5p (miR-186), Sirtuin-3 (SIRT3), Peroxisome Proliferator Activator Receptor alpha (PPAR-α), and Hypoxia-Inducible Factor-1 (HIF-1) in the development of TMX resistance in BC patients and to correlate this axis with tumor progression. Interestingly, this will be the first time to explore epigenetic regulation of this axis in BC.
Collapse
Affiliation(s)
- Hany N Azzam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Marwa O El-Derany
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Sara A Wahdan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Reham M Faheim
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Gouda K Helal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
- Preclinical & Translational Research Center, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
4
|
Ouyang J, Sheng Y, Wang W. Recent Advances of Studies on Cell-Penetrating Peptides Based on Molecular Dynamics Simulations. Cells 2022; 11:cells11244016. [PMID: 36552778 PMCID: PMC9776715 DOI: 10.3390/cells11244016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
With the ability to transport cargo molecules across cell membranes with low toxicity, cell-penetrating peptides (CPPs) have become promising candidates for next generation peptide-based drug delivery vectors. Over the past three decades since the first CPP was discovered, a great deal of work has been done on the cellular uptake mechanisms and the applications for the delivery of therapeutic molecules, and significant advances have been made. But so far, we still do not have a precise and unified understanding of the structure-activity relationship of the CPPs. Molecular dynamics (MD) simulations provide a method to reveal peptide-membrane interactions at the atomistic level and have become an effective complement to experiments. In this paper, we review the progress of the MD simulations on CPP-membrane interactions, including the computational methods and technical improvements in the MD simulations, the research achievements in the CPP internalization mechanism, CPP decoration and coupling, and the peptide-induced membrane reactions during the penetration process, as well as the comparison of simulated and experimental results.
Collapse
Affiliation(s)
- Jun Ouyang
- School of Public Courses, Bengbu Medical College, Bengbu 233030, China
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Yuebiao Sheng
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China
- High Performance Computing Center, Nanjing University, Nanjing 210093, China
- Correspondence: (Y.S.); (W.W.)
| | - Wei Wang
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China
- Correspondence: (Y.S.); (W.W.)
| |
Collapse
|
5
|
Barman B, Kushwaha A, Thakur MK. Muscarinic Acetylcholine Receptors-Mediated Activation of PKC Restores the Hippocampal Immediate Early Gene Expression and CREB Phosphorylation in Scopolamine-Induced Amnesic Mice. Mol Neurobiol 2022; 59:5722-5733. [DOI: 10.1007/s12035-022-02940-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 06/23/2022] [Indexed: 10/17/2022]
|
6
|
Harford TJ, Rezaee F, Gupta MK, Bokun V, Naga Prasad SV, Piedimonte G. Respiratory syncytial virus induces β 2-adrenergic receptor dysfunction in human airway smooth muscle cells. Sci Signal 2021; 14:14/685/eabc1983. [PMID: 34074703 DOI: 10.1126/scisignal.abc1983] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Pharmacologic agonism of the β2-adrenergic receptor (β2AR) induces bronchodilation by activating the enzyme adenylyl cyclase to generate cyclic adenosine monophosphate (cAMP). β2AR agonists are generally the most effective strategy to relieve acute airway obstruction in asthmatic patients, but they are much less effective when airway obstruction in young patients is triggered by infection with respiratory syncytial virus (RSV). Here, we investigated the effects of RSV infection on the abundance and function of β2AR in primary human airway smooth muscle cells (HASMCs) derived from pediatric lung tissue. We showed that RSV infection of HASMCs resulted in proteolytic cleavage of β2AR mediated by the proteasome. RSV infection also resulted in β2AR ligand-independent activation of adenylyl cyclase, leading to reduced cAMP synthesis compared to that in uninfected control cells. Last, RSV infection caused stronger airway smooth muscle cell contraction in vitro due to increased cytosolic Ca2+ concentrations. Thus, our results suggest that RSV infection simultaneously induces loss of functional β2ARs and activation of multiple pathways favoring airway obstruction in young patients, with the net effect of counteracting β2AR agonist-induced bronchodilation. These findings not only provide a potential mechanism for the reported lack of clinical efficacy of β2AR agonists for treating virus-induced wheezing but also open the path to developing more precise therapeutic strategies.
Collapse
Affiliation(s)
- Terri J Harford
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Fariba Rezaee
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Manveen K Gupta
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Vladimir Bokun
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Sathyamangla V Naga Prasad
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Giovanni Piedimonte
- Departments of Pediatrics, Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
7
|
Abdullah CS, Aishwarya R, Alam S, Morshed M, Remex NS, Nitu S, Kolluru GK, Traylor J, Miriyala S, Panchatcharam M, Hartman B, King J, Bhuiyan MAN, Chandran S, Woolard MD, Yu X, Goeders NE, Dominic P, Arnold CL, Stokes K, Kevil CG, Orr AW, Bhuiyan MS. Methamphetamine induces cardiomyopathy by Sigmar1 inhibition-dependent impairment of mitochondrial dynamics and function. Commun Biol 2020; 3:682. [PMID: 33203971 PMCID: PMC7673131 DOI: 10.1038/s42003-020-01408-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Methamphetamine-associated cardiomyopathy is the leading cause of death linked with illicit drug use. Here we show that Sigmar1 is a therapeutic target for methamphetamine-associated cardiomyopathy and defined the molecular mechanisms using autopsy samples of human hearts, and a mouse model of "binge and crash" methamphetamine administration. Sigmar1 expression is significantly decreased in the hearts of human methamphetamine users and those of "binge and crash" methamphetamine-treated mice. The hearts of methamphetamine users also show signs of cardiomyopathy, including cellular injury, fibrosis, and enlargement of the heart. In addition, mice expose to "binge and crash" methamphetamine develop cardiac hypertrophy, fibrotic remodeling, and mitochondrial dysfunction leading to contractile dysfunction. Methamphetamine treatment inhibits Sigmar1, resulting in inactivation of the cAMP response element-binding protein (CREB), decreased expression of mitochondrial fission 1 protein (FIS1), and ultimately alteration of mitochondrial dynamics and function. Therefore, Sigmar1 is a viable therapeutic agent for protection against methamphetamine-associated cardiomyopathy.
Collapse
Affiliation(s)
- Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Richa Aishwarya
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Shafiul Alam
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Mahboob Morshed
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Sadia Nitu
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Gopi K Kolluru
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - James Traylor
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Sumitra Miriyala
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Manikandan Panchatcharam
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Brandon Hartman
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Judy King
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | | | - Sunitha Chandran
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Matthew D Woolard
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Xiuping Yu
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Nicholas E Goeders
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Paari Dominic
- Department of Medicine, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Connie L Arnold
- Department of Medicine, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Karen Stokes
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Christopher G Kevil
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - A Wayne Orr
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Md Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA.
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA.
| |
Collapse
|
8
|
Down-Regulated CUEDC2 Increases GDNF Expression by Stabilizing CREB Through Reducing Its Ubiquitination in Glioma. Neurochem Res 2020; 45:2915-2925. [PMID: 33125618 DOI: 10.1007/s11064-020-03140-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 12/15/2022]
Abstract
Abnormally high expression of glial cell line-derived neurotrophic factor (GDNF) derived from glioma cells has essential impacts on gliomagenesis and development, but the molecular basis underlying increased GDNF expression in glioma cells remain unclear. This work aimed to study the molecular mechanisms that may explain the accumulation of GDNF in glioma. Firstly, we observed that cAMP response element-binding protein (CREB), known as an important transcription factor for binding of GDNF promoter region, was highly expressed with an apparent accumulation into the nucleus of glioma cells, which may contribute to the transcription of GDNF. Secondly, CUE domain-containing protein 2 (CUEDC2), a ubiquitin-regulated protein, could increase the amount of binding between the E3 ligase tripartite motif-containing 21 (TRIM21) and CREB and affect the CREB level. Like our previous study, it showed that there was a significantly down-regulation of CUEDC2 in glioma. Finally, our data suggest that GDNF expression is indirectly regulated by transcription factor ubiquitination. Indeed, down-regulation of CUEDC2, decreased the ubiquitination and degradation of CREB, which was associated to high levels of GDNF. Furthermore, abundant CREB involved in the binding to the GDNF promoter region contributes to GDNF high expression in glioma cells. Collectively, it was verified the GDNF expression was affected by CREB ubiquitination regulated by CUEDC2 level.
Collapse
|
9
|
Steven A, Friedrich M, Jank P, Heimer N, Budczies J, Denkert C, Seliger B. What turns CREB on? And off? And why does it matter? Cell Mol Life Sci 2020; 77:4049-4067. [PMID: 32347317 PMCID: PMC7532970 DOI: 10.1007/s00018-020-03525-8] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/21/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022]
Abstract
Altered expression and function of the transcription factor cyclic AMP response-binding protein (CREB) has been identified to play an important role in cancer and is associated with the overall survival and therapy response of tumor patients. This review focuses on the expression and activation of CREB under physiologic conditions and in tumors of distinct origin as well as the underlying mechanisms of CREB regulation by diverse stimuli and inhibitors. In addition, the clinical relevance of CREB is summarized, including its use as a prognostic and/or predictive marker as well as a therapeutic target.
Collapse
Affiliation(s)
- André Steven
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Michael Friedrich
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Paul Jank
- Institute of Pathology, Philipps University Marburg, 35043, Marburg, Germany
| | - Nadine Heimer
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Jan Budczies
- Institute of Pathology, University Clinic Heidelberg, 69120, Heidelberg, Germany
| | - Carsten Denkert
- Institute of Pathology, Philipps University Marburg, 35043, Marburg, Germany
| | - Barbara Seliger
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany.
| |
Collapse
|
10
|
Protein phosphatase 1 in tumorigenesis: is it worth a closer look? Biochim Biophys Acta Rev Cancer 2020; 1874:188433. [PMID: 32956763 DOI: 10.1016/j.bbcan.2020.188433] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/26/2020] [Accepted: 09/12/2020] [Indexed: 02/06/2023]
Abstract
Cancer cells take advantage of signaling cascades to meet their requirements for sustained growth and survival. Cell signaling is tightly controlled by reversible protein phosphorylation mechanisms, which require the counterbalanced action of protein kinases and protein phosphatases. Imbalances on this system are associated with cancer development and progression. Protein phosphatase 1 (PP1) is one of the most relevant protein phosphatases in eukaryotic cells. Despite the widely recognized involvement of PP1 in key biological processes, both in health and disease, its relevance in cancer has been largely neglected. Here, we provide compelling evidence that support major roles for PP1 in tumorigenesis.
Collapse
|
11
|
Khan N, Chen X, Geiger JD. Role of Divalent Cations in HIV-1 Replication and Pathogenicity. Viruses 2020; 12:E471. [PMID: 32326317 PMCID: PMC7232465 DOI: 10.3390/v12040471] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/09/2020] [Accepted: 04/18/2020] [Indexed: 12/22/2022] Open
Abstract
Divalent cations are essential for life and are fundamentally important coordinators of cellular metabolism, cell growth, host-pathogen interactions, and cell death. Specifically, for human immunodeficiency virus type-1 (HIV-1), divalent cations are required for interactions between viral and host factors that govern HIV-1 replication and pathogenicity. Homeostatic regulation of divalent cations' levels and actions appear to change as HIV-1 infection progresses and as changes occur between HIV-1 and the host. In people living with HIV-1, dietary supplementation with divalent cations may increase HIV-1 replication, whereas cation chelation may suppress HIV-1 replication and decrease disease progression. Here, we review literature on the roles of zinc (Zn2+), iron (Fe2+), manganese (Mn2+), magnesium (Mg2+), selenium (Se2+), and copper (Cu2+) in HIV-1 replication and pathogenicity, as well as evidence that divalent cation levels and actions may be targeted therapeutically in people living with HIV-1.
Collapse
Affiliation(s)
| | | | - Jonathan D. Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA; (N.K.); (X.C.)
| |
Collapse
|
12
|
Vashisht A, Bach SV, Fetterhoff D, Morgan JW, McGee M, Hegde AN. Proteasome limits plasticity-related signaling to the nucleus in the hippocampus. Neurosci Lett 2018; 687:31-36. [PMID: 30219486 DOI: 10.1016/j.neulet.2018.09.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/21/2018] [Accepted: 09/10/2018] [Indexed: 01/22/2023]
Abstract
Proteolysis by the ubiquitin-proteasome pathway has pleiotropic effects on both induction and maintenance of long-term synaptic plasticity. In this study, we examined the effect of proteasome inhibition on signaling to the nucleus during late-phase long-term potentiation. When a subthreshold L-LTP induction protocol was used, proteasome inhibition led to a significant increase in phosphorylated CREB (pCREB) in the nucleus. Inhibitors of cAMP-dependent protein kinase/protein kinase A, extracellular signal-regulated kinase and cGMP-dependent protein kinase/protein kinase G all blocked the proteasome-inhibition-mediated increase in nuclear pCREB after subthreshold stimulation. These results lay the groundwork for understanding a novel role for the proteasome in limiting signaling to the nucleus in the absence of adequate synaptic stimulation.
Collapse
Affiliation(s)
- Anirudh Vashisht
- Department of Neurobiology and Anatomy & Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, 27157, USA; Department of Biological and Environmental Sciences, Georgia College and State University, Milledgeville, GA, 31061, USA
| | - Svitlana V Bach
- Department of Neurobiology and Anatomy & Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Dustin Fetterhoff
- Department of Neurobiology and Anatomy & Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - James W Morgan
- Department of Neurobiology and Anatomy & Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Maria McGee
- Plastic and Reconstructive Surgery Research, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Ashok N Hegde
- Department of Neurobiology and Anatomy & Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC, 27157, USA; Department of Biological and Environmental Sciences, Georgia College and State University, Milledgeville, GA, 31061, USA.
| |
Collapse
|
13
|
Mishra R, Upadhyay A, Prajapati VK, Mishra A. Proteasome-mediated proteostasis: Novel medicinal and pharmacological strategies for diseases. Med Res Rev 2018; 38:1916-1973. [DOI: 10.1002/med.21502] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 03/13/2018] [Accepted: 04/04/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Ribhav Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan India
| | - Arun Upadhyay
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan India
| | - Vijay Kumar Prajapati
- Department of Biochemistry; School of Life Sciences; Central University of Rajasthan; Rajasthan India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan India
| |
Collapse
|
14
|
Steven A, Leisz S, Sychra K, Hiebl B, Wickenhauser C, Mougiakakos D, Kiessling R, Denkert C, Seliger B. Hypoxia-mediated alterations and their role in the HER-2/neuregulated CREB status and localization. Oncotarget 2018; 7:52061-52084. [PMID: 27409833 PMCID: PMC5239535 DOI: 10.18632/oncotarget.10474] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/17/2016] [Indexed: 01/16/2023] Open
Abstract
The cAMP-responsive element-binding protein (CREB) is involved in the tumorigenicity of HER-2/neu-overexpressing murine and human tumor cells, but a link between the HER-2/neu-mediated CREB activation, its posttranslational modification and localization and changes in the cellular metabolism, due to an altered (tumor) microenvironment remains to be established. The present study demonstrated that shRNA-mediated silencing of CREB in HER-2/neu-transformed cells resulted in decreased tumor formation, which was associated with reduced angiogenesis, but increased necrotic and hypoxic areas in the tumor. Hypoxia induced pCREBSer133, but not pCREBSer121 expression in HER-2/neu-transformed cells. This was accompanied by upregulation of the hypoxia-inducible genes GLUT1 and VEGF, increased cell migration and matrix metalloproteinase-mediated invasion. Treatment of HER-2/neu+ cells with signal transduction inhibitors targeting in particular HER-2/neu was able to revert hypoxia-controlled CREB activation. In addition to changes in the phosphorylation, hypoxic response of HER-2/neu+ cells caused a transient ubiquitination and SUMOylation as well as a co-localization of nuclear CREB to the mitochondrial matrix. A mitochondrial localization of CREB was also demonstrated in hypoxic areas of HER-2/neu+ mammary carcinoma lesions. This was accompanied by an altered gene expression pattern, activity and metabolism of mitochondria leading to an increased respiratory rate, oxidative phosphorylation and mitochondrial membrane potential and consequently to an enhanced apoptosis and reduced cell viability. These data suggest that the HER-2/neu-mediated CREB activation caused by a hypoxic tumor microenvironment contributes to the neoplastic phenotype of HER-2/neu+ cells at various levels.
Collapse
Affiliation(s)
- André Steven
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Sandra Leisz
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | | | - Bernhard Hiebl
- Centre for Basic Medical Research, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Claudia Wickenhauser
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
15
|
Steven A, Seliger B. Control of CREB expression in tumors: from molecular mechanisms and signal transduction pathways to therapeutic target. Oncotarget 2018; 7:35454-65. [PMID: 26934558 PMCID: PMC5085243 DOI: 10.18632/oncotarget.7721] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 01/26/2016] [Indexed: 12/11/2022] Open
Abstract
The cyclic AMP response element binding (CREB) protein has pleiotropic activities in physiologic processes. Due to its central position downstream of many growth signaling pathways CREB has the ability to influence cell survival, growth and differentiation of normal, but also of tumor cells suggesting an oncogenic potential of CREB. Indeed, increased CREB expression and activation is associated with tumor progression, chemotherapy resistance and reduced patients' survival. We summarize here the different cellular functions of CREB in tumors of distinct histology as well as its use as potential prognostic marker. In addition, the underlying molecular mechanisms to achieve constitutive activation of CREB including structural alterations, such as gene amplification and chromosomal translocation, and deregulation, which could occur at the transcriptional, post-transcriptional and post-translational level, will be described. Since downregulation of CREB by different strategies resulted in inhibition of cell proliferation, invasion and induction of apoptosis, the role of CREB as a promising target for cancer therapy will be also discussed.
Collapse
Affiliation(s)
- André Steven
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
16
|
Wang SH, Wang SC, Chen PC, Wang ST, Liu YW. Induction of cyclooxygenase-2 gene by Candida albicans through EGFR, ERK, and p38 pathways in human urinary epithelium. Med Mycol 2017; 55:314-322. [PMID: 27664170 DOI: 10.1093/mmy/myw082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 06/15/2016] [Indexed: 12/26/2022] Open
Abstract
In the present data, we found that Candida albicans (C. albicans) caused bladder epithelial cell morphology alteration, cell damage, and inflammatory responses, including cyclooxygenase-2 (COX-2) gene and protein expression as well as prostaglandin E2 accumulation. In addition, the molecular pathway underlying C. albicans-induced urothelial COX-2 gene expression was examined. Among MAPK pathways, phosphorylation of ERK1/2, p38, and JNK each increased following C. albicans infection for 12 h. However, C. albicans-induced COX-2 protein expression was inhibited by specific inhibitors of ERK and p38 (U0126 and SB203580) but not by JNK inhibitor SP600125. Additional evidence came from the increased amount of phosphorylated RSK that is the mutual downstream molecule of ERK1/2 and p38. Furthermore, phosphorylation of RSK protein was reduced by the ERK and p38 inhibitor, suggesting that the urothelial COX-2 gene was induced majorly though the ERK/p38-RSK pathway by C. albicans infection. We also found transcription factor CREB-1 showed increased binding to the COX-2 gene promoter by chromatin immunoprecipitation assay. Next, we used receptor inhibitors including Toll-like receptor (TLR)-Myd88 inhibitor ST2825, Dectin-Syk inhibitor Syk inhibitor, and epidermal growth factor receptor (EGFR) inhibitor PD168393 to identify which one was the main target associated with C. albicans binding. The results revealed that it was EGFR, recognized by C. albicans, that mostly mediated the ERK/p38-RSK pathway activation to induce COX-2 gene expression, but this was not the case for TLRs and Dectin receptors. In summary, these results demonstrated the EGFR-ERK/p38-RSK-CREB-1 pathway was involved significantly in the C. albicans-induced COX-2 expression in human urothelium.
Collapse
Affiliation(s)
- Shao-Hung Wang
- Department of Microbiology, Immunology and Biopharmaceuticals; College of Life Sciences, National Chiayi University, Chiayi 600, Taiwan
| | - Shou-Chieh Wang
- Division of Nephrology, Department of Internal Medicine, Kuang Tien General Hospital, Taichung 437, Taiwan.,Department of Food Science; College of Life Sciences, National Chiayi University, Chiayi 600, Taiwan
| | - Pei-Ching Chen
- Department of Microbiology, Immunology and Biopharmaceuticals; College of Life Sciences, National Chiayi University, Chiayi 600, Taiwan
| | - Shou-Tsung Wang
- Department of Food Science; College of Life Sciences, National Chiayi University, Chiayi 600, Taiwan
| | - Yi-Wen Liu
- Department of Microbiology, Immunology and Biopharmaceuticals; College of Life Sciences, National Chiayi University, Chiayi 600, Taiwan
| |
Collapse
|
17
|
Malki K, Tosto MG, Mouriño‐Talín H, Rodríguez‐Lorenzo S, Pain O, Jumhaboy I, Liu T, Parpas P, Newman S, Malykh A, Carboni L, Uher R, McGuffin P, Schalkwyk LC, Bryson K, Herbster M. Highly polygenic architecture of antidepressant treatment response: Comparative analysis of SSRI and NRI treatment in an animal model of depression. Am J Med Genet B Neuropsychiatr Genet 2017; 174:235-250. [PMID: 27696737 PMCID: PMC5434854 DOI: 10.1002/ajmg.b.32494] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 08/15/2016] [Indexed: 11/12/2022]
Abstract
Response to antidepressant (AD) treatment may be a more polygenic trait than previously hypothesized, with many genetic variants interacting in yet unclear ways. In this study we used methods that can automatically learn to detect patterns of statistical regularity from a sparsely distributed signal across hippocampal transcriptome measurements in a large-scale animal pharmacogenomic study to uncover genomic variations associated with AD. The study used four inbred mouse strains of both sexes, two drug treatments, and a control group (escitalopram, nortriptyline, and saline). Multi-class and binary classification using Machine Learning (ML) and regularization algorithms using iterative and univariate feature selection methods, including InfoGain, mRMR, ANOVA, and Chi Square, were used to uncover genomic markers associated with AD response. Relevant genes were selected based on Jaccard distance and carried forward for gene-network analysis. Linear association methods uncovered only one gene associated with drug treatment response. The implementation of ML algorithms, together with feature reduction methods, revealed a set of 204 genes associated with SSRI and 241 genes associated with NRI response. Although only 10% of genes overlapped across the two drugs, network analysis shows that both drugs modulated the CREB pathway, through different molecular mechanisms. Through careful implementation and optimisations, the algorithms detected a weak signal used to predict whether an animal was treated with nortriptyline (77%) or escitalopram (67%) on an independent testing set. The results from this study indicate that the molecular signature of AD treatment may include a much broader range of genomic markers than previously hypothesized, suggesting that response to medication may be as complex as the pathology. The search for biomarkers of antidepressant treatment response could therefore consider a higher number of genetic markers and their interactions. Through predominately different molecular targets and mechanisms of action, the two drugs modulate the same Creb1 pathway which plays a key role in neurotrophic responses and in inflammatory processes. © 2016 The Authors. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Karim Malki
- King's College LondonMRC SocialGenetic and Developmental Psychiatry Centre at the Institute of PsychiatryPsychology and Neuroscience (IOPPN)LondonUnited Kingdom
| | - Maria Grazia Tosto
- King's College LondonMRC SocialGenetic and Developmental Psychiatry Centre at the Institute of PsychiatryPsychology and Neuroscience (IOPPN)LondonUnited Kingdom,LCIBGTomsk State UniversityTomskRussia
| | | | | | - Oliver Pain
- BirkbeckUniversity of LondonUnited Kingdom,London School of Hygiene & Tropical MedicineUnited Kingdom
| | - Irfan Jumhaboy
- King's College LondonMRC SocialGenetic and Developmental Psychiatry Centre at the Institute of PsychiatryPsychology and Neuroscience (IOPPN)LondonUnited Kingdom
| | - Tina Liu
- Department of Computer Science Imperial College LondonUnited Kingdom
| | - Panos Parpas
- Department of Computer Science Imperial College LondonUnited Kingdom
| | - Stuart Newman
- King's College LondonMRC SocialGenetic and Developmental Psychiatry Centre at the Institute of PsychiatryPsychology and Neuroscience (IOPPN)LondonUnited Kingdom
| | | | - Lucia Carboni
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum University of BolognaBolognaItaly
| | - Rudolf Uher
- King's College LondonMRC SocialGenetic and Developmental Psychiatry Centre at the Institute of PsychiatryPsychology and Neuroscience (IOPPN)LondonUnited Kingdom,Department of PsychiatryDalhousie UniversityHalifaxNova ScotiaCanada
| | - Peter McGuffin
- King's College LondonMRC SocialGenetic and Developmental Psychiatry Centre at the Institute of PsychiatryPsychology and Neuroscience (IOPPN)LondonUnited Kingdom
| | | | - Kevin Bryson
- Department of Computer ScienceUCLLondonUnited Kingdom
| | - Mark Herbster
- Department of Computer ScienceUCLLondonUnited Kingdom
| |
Collapse
|
18
|
Increased iron export by ferroportin induces restriction of HIV-1 infection in sickle cell disease. Blood Adv 2016; 1:170-183. [PMID: 28203649 DOI: 10.1182/bloodadvances.2016000745] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The low incidence of HIV-1 infection in patients with sickle cell disease (SCD) and inhibition of HIV-1 replication in vitro under the conditions of low intracellular iron or heme treatment suggests a potential restriction of HIV-1 infection in SCD. We investigated HIV-1 ex vivo infection of SCD peripheral blood mononuclear cells (PBMCs) and found that HIV-1 replication was inhibited at the level of reverse transcription (RT) and transcription. We observed increased expression of heme and iron-regulated genes, previously shown to inhibit HIV-1, including ferroportin, IKBα, HO-1, p21, and SAM domain and HD domain-containing protein 1 (SAMHD1). HIV-1 inhibition was less pronounced in hepcidin-treated SCD PBMCs and more pronounced in the iron or iron chelators treated, suggesting a key role of iron metabolism. In SCD PBMCs, labile iron levels were reduced and protein levels of ferroportin, HIF-1α, IKBα, and HO-1 were increased. Hemin treatment induced ferroportin expression and inhibited HIV-1 in THP-1 cells, mimicking the HIV-1 inhibition in SCD PBMCs, especially as hepcidin similarly prevented HIV-1 inhibition. In THP-1 cells with knocked down ferroportin, IKBα, or HO-1 genes but not HIF-1α or p21, HIV-1 was not inhibited by hemin. Activity of SAMHD1-regulatory CDK2 was decreased, and SAMHD1 phosphorylation was reduced in SCD PBMCs and hemin-treated THP-1 cells, suggesting SAMHD1-mediated HIV-1 restriction in SCD. Our findings point to ferroportin as a trigger of HIV-1 restriction in SCD settings, linking reduced intracellular iron levels to the inhibition of CDK2 activity, reduction of SAMHD1 phosphorylation, increased IKBα expression, and inhibition of HIV-1 RT and transcription.
Collapse
|
19
|
Cummins EP, Keogh CE. Respiratory gases and the regulation of transcription. Exp Physiol 2016; 101:986-1002. [DOI: 10.1113/ep085715] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 05/23/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Eoin P. Cummins
- School of Medicine; University College Dublin; Belfield 4 Dublin Ireland
| | - Ciara E. Keogh
- School of Medicine; University College Dublin; Belfield 4 Dublin Ireland
| |
Collapse
|
20
|
Tang Q, Huang W, Guan J, Jin L, Che T, Fu Y, Hu Y, Tian S, Wang D, Jiang Z, Li X, Li M. Transcriptomic analysis provides insight into high-altitude acclimation in domestic goats. Gene 2015; 567:208-16. [PMID: 25958351 DOI: 10.1016/j.gene.2015.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/27/2015] [Accepted: 05/01/2015] [Indexed: 10/23/2022]
Abstract
Domestic goats are distributed in a wide range of habitats and have acclimated to their local environmental conditions. To investigate the gene expression changes of goats that are induced by high altitude stress, we performed RNA-seq on 27 samples from the three hypoxia-sensitive tissues (heart, lung, and skeletal muscle) in three indigenous populations from distinct altitudes (600 m, 2000 m, and 3000 m). We generated 129Gb of high-quality sequencing data (~4Gb per sample) and catalogued the expression profiles of 12,421 annotated hircine genes in each sample. The analysis showed global similarities and differences of high-altitude transcriptomes among populations and tissues as well as revealed that the heart underwent the most high-altitude induced expression changes. We identified numerous differentially expressed genes that exhibited distinct expression patterns, and nonsynonymous single nucleotide variant-containing genes that were highly differentiated between the high- and low-altitude populations. These genes have known or potential roles in hypoxia response and were enriched in functional gene categories potentially responsible for high-altitude stress. Therefore, they are appealing candidates for further investigation of the gene expression and associated regulatory mechanisms related to high-altitude acclimation.
Collapse
Affiliation(s)
- Qianzi Tang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Wenyao Huang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiuqiang Guan
- Sichuan Academy of Grassland Science, Chengdu 611731, China
| | - Long Jin
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Tiandong Che
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuhua Fu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Lab of Animal Genetics, Breeding and Reproduction of Ministry Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Yaodong Hu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shilin Tian
- Novogene Bioinformatics Institute, Beijing 100083, China
| | - Dawei Wang
- Novogene Bioinformatics Institute, Beijing 100083, China
| | - Zhi Jiang
- Novogene Bioinformatics Institute, Beijing 100083, China
| | - Xuewei Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingzhou Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
21
|
Felsenberg J, Dyck Y, Feige J, Ludwig J, Plath JA, Froese A, Karrenbrock M, Nölle A, Heufelder K, Eisenhardt D. Differences in long-term memory stability and AmCREB level between forward and backward conditioned honeybees (Apis mellifera). Front Behav Neurosci 2015; 9:91. [PMID: 25964749 PMCID: PMC4410603 DOI: 10.3389/fnbeh.2015.00091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/30/2015] [Indexed: 12/25/2022] Open
Abstract
In classical conditioning a predictive relationship between a neutral stimulus (conditioned stimulus; CS) and a meaningful stimulus (unconditioned stimulus; US) is learned when the CS precedes the US. In backward conditioning the sequence of the stimuli is reversed. In this situation animals might learn that the CS signals the end or the absence of the US. In honeybees 30 min and 24 h following backward conditioning a memory for the excitatory and inhibitory properties of the CS could be retrieved, but it remains unclear whether a late long-term memory is formed that can be retrieved 72 h following backward conditioning. Here we examine this question by studying late long-term memory formation in forward and backward conditioning of the proboscis extension response (PER). We report a difference in the stability of memory formed upon forward and backward conditioning with the same number of conditioning trials. We demonstrate a transcription-dependent memory 72 h after forward conditioning but do not observe a 72 h memory after backward conditioning. Moreover we find that protein degradation is differentially involved in memory formation following these two conditioning protocols. We report differences in the level of a transcription factor, the cAMP response element binding protein (CREB) known to induce transcription underlying long-term memory formation, following forward and backward conditioning. Our results suggest that these alterations in CREB levels might be regulated by the proteasome. We propose that the differences observed are due to the sequence of stimulus presentation between forward and backward conditioning and not to differences in the strength of the association of both stimuli.
Collapse
Affiliation(s)
- Johannes Felsenberg
- FB Biologie, Pharmazie, Chemie, Institut für Biologie, Neurobiologie, Freie Universität Berlin Berlin, Germany
| | - Yan Dyck
- FB Biologie, Pharmazie, Chemie, Institut für Biologie, Neurobiologie, Freie Universität Berlin Berlin, Germany
| | - Janina Feige
- FB Biologie, Pharmazie, Chemie, Institut für Biologie, Neurobiologie, Freie Universität Berlin Berlin, Germany
| | - Jenny Ludwig
- FB Biologie, Pharmazie, Chemie, Institut für Biologie, Neurobiologie, Freie Universität Berlin Berlin, Germany
| | - Jenny Aino Plath
- FB Biologie, Pharmazie, Chemie, Institut für Biologie, Neurobiologie, Freie Universität Berlin Berlin, Germany
| | - Anja Froese
- FB Biologie, Pharmazie, Chemie, Institut für Biologie, Neurobiologie, Freie Universität Berlin Berlin, Germany
| | - Melanie Karrenbrock
- FB Biologie, Pharmazie, Chemie, Institut für Biologie, Neurobiologie, Freie Universität Berlin Berlin, Germany
| | - Anna Nölle
- FB Biologie, Pharmazie, Chemie, Institut für Biologie, Neurobiologie, Freie Universität Berlin Berlin, Germany
| | - Karin Heufelder
- FB Biologie, Pharmazie, Chemie, Institut für Biologie, Neurobiologie, Freie Universität Berlin Berlin, Germany
| | - Dorothea Eisenhardt
- FB Biologie, Pharmazie, Chemie, Institut für Biologie, Neurobiologie, Freie Universität Berlin Berlin, Germany
| |
Collapse
|
22
|
Yun CY, You ST, Kim JH, Chung JH, Han SB, Shin EY, Kim EG. p21-activated kinase 4 critically regulates melanogenesis via activation of the CREB/MITF and β-catenin/MITF pathways. J Invest Dermatol 2015; 135:1385-1394. [PMID: 25560280 DOI: 10.1038/jid.2014.548] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 12/01/2014] [Accepted: 12/16/2014] [Indexed: 01/14/2023]
Abstract
p21-activated kinase 4 (PAK4) regulates a wide range of cellular events, including cytoskeletal remodeling, cell growth, and survival. Our previous study identified PAK4 as a key regulator of cAMP-response element-binding protein (CREB) that acts upstream of microphthalmia-associated transcription factor (MITF), a master transcription factor in melanogenesis. We therefore investigated the role of PAK4 in melanogenesis. Melanocytes express both PAK2 and PAK4 isoforms, but only RNA interference knockdown of PAK4 significantly influenced α-melanocyte-stimulating hormone (α-MSH)-induced melanogenesis in B16 melanoma cells. Consistent with this result, PAK4 inhibition by PF3758309, a potent ATP-competitive inhibitor of PAKs, suppressed not only α-MSH-induced melanogenesis in B16 melanoma and human epithelial melanocyte cells but also UVB-induced melanogenesis in the skin of melanin-possessing hairless mice (HRM-2) in a dose-dependent manner. Inhibition of PAK4 over several days markedly decreased the levels of CREB, MITF, and tyrosinase in both HRM-2 mice and B16 melanoma cells. Moreover, PAK4 knockdown and inhibition suppressed α-MSH-stimulated β-catenin phosphorylation at serine 675 (S675) but enhanced phosphorylation at S33/37, an indicator for ubiquitination-dependent proteolysis. Together, our results provide evidence that PAK4 promotes α-MSH/UVB-induced melanogenesis via the CREB and Wnt/β-catenin signaling pathways and suggest that PAK4 may be a potential therapeutic target in pigmentation disorders.
Collapse
Affiliation(s)
- Cheong-Yong Yun
- Department of Biochemistry and Medical Research Center, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Soon-Tae You
- Department of Biochemistry and Medical Research Center, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Jin-Hwa Kim
- R&D Center, Hanbul Cosmetics Co. Ltd, Chungbuk, Korea.
| | - Jin H Chung
- Department of Dermatology, Seoul National University, Seoul, Korea
| | - Sang-Bae Han
- College of Pharmacy and CBITRC, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Eun-Young Shin
- Department of Biochemistry and Medical Research Center, College of Medicine, Chungbuk National University, Cheongju, Korea.
| | - Eung-Gook Kim
- Department of Biochemistry and Medical Research Center, College of Medicine, Chungbuk National University, Cheongju, Korea
| |
Collapse
|
23
|
Zhang S, Zhang Y, Jiang S, Liu Y, Huang L, Zhang T, Lu G, Gong K, Ji X, Shao G. The effect of hypoxia preconditioning on DNA methyltransferase and PP1γ in hippocampus of hypoxia preconditioned mice. High Alt Med Biol 2014; 15:483-90. [PMID: 25531462 DOI: 10.1089/ham.2014.1042] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
It is well known that hypoxia preconditioning can increase hypoxic tolerance by changing the expressions of some genes in the brain. DNA methylation is important for regulating gene expression and is catalyzed by DNA methyltransferase (DNMT), an enzyme that is abundant in the brain. However, the impact of hypoxia preconditioning on DNA methylation remains unknown. In the current study, mice were randomly divided into three groups: blank control group with no exposure to hypoxia (H0), the hypoxia control group exposed to hypoxia once (H1), and the hypoxia preconditioning group exposed to 4 runs of hypoxia (H4). The mRNA and protein levels of three kinds of DNMTs and the activity of total DMNT were measured. Protein phosphatase 1(PP1)γ, which critically regulates neuroprotective pathways in brain, was measured in mRNA and protein activity and promoter methylation. DNMT1 was unchanged in H1 and H4, while DNMT3A and DNMT3B were decreased in H4. The mRNA and protein levels of PP1γ were decreased in H4. However, there was no detectable change in the level of DNA methylation of the promoter of PP1γ (-321 bp to 95 bp). These findings suggest that DNA methylation may have a role in hypoxia neuroprotection, and the change of PP1γ, which did not depend on the change of its promoter (-321 bp to 95bp) DNA methylation, may be involved in neuroprotection.
Collapse
Affiliation(s)
- Shu Zhang
- 1 Biomedicine Research Center and Basic Medical College, The First Affiliated Hospital of BaoTou Medical College , Inner Mongolia, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
|
25
|
Rolewska P, Simm A, Silber RE, Bartling B. Reduced expression level of the cyclic adenosine monophosphate response element-binding protein contributes to lung aging. Am J Respir Cell Mol Biol 2014; 50:201-11. [PMID: 23991634 DOI: 10.1165/rcmb.2013-0057oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Lung aging is associated with morphological and physiological changes in which alterations in transcription factors, including the cyclic adenosine monophosphate response element-binding protein (CREB), could play a role. We studied CREB in lung tissue from mice at different ages and in response to known age-related factors (e.g., cellular senescence and matrix modifications with advanced glycation end-products [AGEs]). Our study shows that protein but not mRNA levels of CREB are reduced in the lungs of old mice. CREB reduction was also observed in senescent human lung fibroblasts (WI-38, LuFi) and human lung epithelial cells (A549) cultured on AGE-modified collagen matrix. Reduction of CREB protein is partially based on pre- and posttranslational modifications as exhibited by an increase in the CREB-regulating microRNA 34b and CREB ubiquitination. Permanent down-regulation of CREB in lung cells impaired cell proliferation and viability and increased the number of cells with senescence-associated β-galactosidase activity. CREB down-regulation was accompanied by the reduced expression of 165 genes in WI-38 fibroblasts and A549 epithelial cells, of which 15 genes showed a reduced expression in lung tissues of old mice. The CREB-dependent reduction in RAB27A coding for the Ras-related protein Rab27A and IGFBP3 coding for the insulin-like growth factor-binding protein 3 has been confirmed for aged lung tissue, senescent fibroblasts, and lung epithelial cells on AGE-modified collagen. Our data demonstrate that the reduced protein expression of CREB might play a significant role in lung aging by modifying the transcription of RAB27A, IGFBP3, and other target genes.
Collapse
Affiliation(s)
- Paulina Rolewska
- 1 Department of Cardio-Thoracic Surgery, University Hospital Halle/Saale, Martin Luther University Halle-Wittenberg, Halle/Saale, Germany
| | | | | | | |
Collapse
|
26
|
Li L, Howell K, Sands M, Banahan M, Frohlich S, Rowan SC, Neary R, Ryan D, McLoughlin P. The α and Δ isoforms of CREB1 are required to maintain normal pulmonary vascular resistance. PLoS One 2013; 8:e80637. [PMID: 24349008 PMCID: PMC3857174 DOI: 10.1371/journal.pone.0080637] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 10/05/2013] [Indexed: 01/15/2023] Open
Abstract
Chronic hypoxia causes pulmonary hypertension associated with structural alterations in pulmonary vessels and sustained vasoconstriction. The transcriptional mechanisms responsible for these distinctive changes are unclear. We have previously reported that CREB1 is activated in the lung in response to alveolar hypoxia but not in other organs. To directly investigate the role of α and Δ isoforms of CREB1 in the regulation of pulmonary vascular resistance we examined the responses of mice in which these isoforms of CREB1 had been inactivated by gene mutation, leaving only the β isoform intact (CREB(αΔ) mice). Here we report that expression of CREB regulated genes was altered in the lungs of CREB(αΔ) mice. CREB(αΔ) mice had greater pulmonary vascular resistance than wild types, both basally in normoxia and following exposure to hypoxic conditions for three weeks. There was no difference in rho kinase mediated vasoconstriction between CREB(αΔ) and wild type mice. Stereological analysis of pulmonary vascular structure showed characteristic wall thickening and lumen reduction in hypoxic wild-type mice, with similar changes observed in CREB(αΔ). CREB(αΔ) mice had larger lungs with reduced epithelial surface density suggesting increased pulmonary compliance. These findings show that α and Δ isoforms of CREB1 regulate homeostatic gene expression in the lung and that normal activity of these isoforms is essential to maintain low pulmonary vascular resistance in both normoxic and hypoxic conditions and to maintain the normal alveolar structure. Interventions that enhance the actions of α and Δ isoforms of CREB1 warrant further investigation in hypoxic lung diseases.
Collapse
Affiliation(s)
- Lili Li
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
| | - Katherine Howell
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
| | - Michelle Sands
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
| | - Mark Banahan
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
| | - Stephen Frohlich
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
- Department of Anaesthesia and Critical Care, St Vincent's University Hospital, Dublin, Ireland
| | - Simon C. Rowan
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
| | - Roisín Neary
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
| | - Donal Ryan
- Department of Anaesthesia and Critical Care, St Vincent's University Hospital, Dublin, Ireland
| | - Paul McLoughlin
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
| |
Collapse
|
27
|
Zhang J, Yin JCP, Wesley CS. From Drosophila development to adult: clues to Notch function in long-term memory. Front Cell Neurosci 2013; 7:222. [PMID: 24312012 PMCID: PMC3836050 DOI: 10.3389/fncel.2013.00222] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 11/03/2013] [Indexed: 12/26/2022] Open
Abstract
Notch is a cell surface receptor that is well known to mediate inter-cellular communication during animal development. Data in the field indicate that it is also involved in the formation of long-term memory (LTM) in the fully developed adults and in memory loss upon neurodegeneration. Our studies in the model organism Drosophila reveal that a non-canonical Notch-protein kinase C activity that plays critical roles in embryonic development also regulates cyclic-AMP response element binding protein during LTM formation in adults. Here we present a perspective on how the various known features of Notch function relate to LTM formation and how they might interface with elements of Wingless/Wnt signaling in this process.
Collapse
Affiliation(s)
- Jiabin Zhang
- Neuroscience Training Program, University of Wisconsin-Madison Madison, WI, USA ; Department of Genetics and Medical Genetics, University of Wisconsin-Madison Madison, WI, USA
| | | | | |
Collapse
|
28
|
Wang Y, Hu Z, Liu Z, Chen R, Peng H, Guo J, Chen X, Zhang H. MTOR inhibition attenuates DNA damage and apoptosis through autophagy-mediated suppression of CREB1. Autophagy 2013; 9:2069-86. [PMID: 24189100 DOI: 10.4161/auto.26447] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Hyperactivation of mechanistic target of rapamycin (MTOR) is a common feature of human cancers, and MTOR inhibitors, such as rapamycin, are thus becoming therapeutics in targeting certain cancers. However, rapamycin has also been found to compromise the efficacy of chemotherapeutics to cells with hyperactive MTOR. Here, we show that loss of TSC2 or PTEN enhanced etoposide-induced DNA damage and apoptosis, which was blunted by suppression of MTOR with either rapamycin or RNA interference. cAMP response element-binding protein 1 (CREB1), a nuclear transcription factor that regulates genes involved in survival and death, was positively regulated by MTOR in mouse embryonic fibroblasts (MEFs) and cancer cell lines. Silencing Creb1 expression with siRNA protected MTOR-hyperactive cells from DNA damage-induced apoptosis. Furthermore, loss of TSC2 or PTEN impaired either etoposide or nutrient starvation-induced autophagy, which in turn, leads to CREB1 hyperactivation. We further elucidated an inverse correlation between autophagy activity and CREB1 activity in the kidney tumor tissue obtained from a TSC patient and the mouse livers with hepatocyte-specific knockout of PTEN. CREB1 induced DNA damage and subsequent apoptosis in response to etoposide in autophagy-defective cells. Reactivation of CREB1 or inhibition of autophagy not only improved the efficacy of rapamycin but also alleviated MTOR inhibition-mediated chemoresistance. Therefore, autophagy suppression of CREB1 may underlie the MTOR inhibition-mediated chemoresistance. We suggest that inhibition of MTOR in combination with CREB1 activation may be used in the treatment of cancer caused by an abnormal PI3K-PTEN-AKT-TSC1/2-MTOR signaling pathway. CREB1 activators should potentiate the efficacy of chemotherapeutics in treatment of these cancers.
Collapse
Affiliation(s)
- Ying Wang
- State Key Laboratory of Medical Molecular Biology; Department of Physiology; Institute of Basic Medical Sciences and School of Basic Medicine; Graduate School of Peking Union Medical College; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
GSK3β controls epithelial-mesenchymal transition and tumor metastasis by CHIP-mediated degradation of Slug. Oncogene 2013; 33:3172-82. [PMID: 23851495 DOI: 10.1038/onc.2013.279] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 05/19/2013] [Accepted: 05/19/2013] [Indexed: 12/22/2022]
Abstract
Glycogen synthase kinase 3 beta (GSK3β) is highly inactivated in epithelial cancers and is known to inhibit tumor migration and invasion. The zinc-finger-containing transcriptional repressor, Slug, represses E-cadherin transcription and enhances epithelial-mesenchymal transition (EMT). In this study, we find that the GSK3β-pSer9 level is associated with the expression of Slug in non-small cell lung cancer. GSK3β-mediated phosphorylation of Slug facilitates Slug protein turnover. Proteomic analysis reveals that the carboxyl terminus of Hsc70-interacting protein (CHIP) interacts with wild-type Slug (wtSlug). Knockdown of CHIP stabilizes the wtSlug protein and reduces Slug ubiquitylation and degradation. In contrast, nonphosphorylatable Slug-4SA is not degraded by CHIP. The accumulation of nondegradable Slug may further lead to the repression of E-cadherin expression and promote cancer cell migration, invasion and metastasis. Our findings provide evidence of a de novo GSK3β-CHIP-Slug pathway that may be involved in the progression of metastasis in lung cancer.
Collapse
|
30
|
Nakayama K. cAMP-response element-binding protein (CREB) and NF-κB transcription factors are activated during prolonged hypoxia and cooperatively regulate the induction of matrix metalloproteinase MMP1. J Biol Chem 2013; 288:22584-95. [PMID: 23775082 DOI: 10.1074/jbc.m112.421636] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Responses to low levels of oxygen (hypoxia) are essential to maintain homeostasis. During the hypoxic response, gene expression is altered by various transcription factors. The transcription factor, hypoxia-inducible factor (HIF), plays a central role in the hypoxic response. The α subunit of HIF, which is actively degraded during normoxia, becomes stabilized during hypoxia, which leads to HIF activation. A microarray analysis of HeLa cells showed that expression of matrix metalloproteinase 1 (MMP1) was markedly induced during prolonged hypoxia. CREB and NF-κB binding sites were identified in the MMP1 promoter region between 1945 and 1896 nucleotides upstream of the transcription start site. Assays with luciferase reporters demonstrated that HIF activity was induced during the early phase of hypoxia, whereas CREB and NF-κB were activated during the later (prolonged) phase. Depletion of CREB and/or NF-κB reduced MMP1 induction during prolonged hypoxia both at the mRNA and protein levels. A chromatin immunoprecipitation assay demonstrated binding of CREB and NF-κB to the MMP1 promoter. Finally, cell migration and invasion on a collagen matrix and pulmonary metastasis in nude mice were inhibited after depletion of CREB and NF-κB in MDA-MB-231 cells. Taken together, these results suggest that the cooperative action of CREB and NF-κB plays an important role to induce MMP1 expression during prolonged hypoxia and regulates cell migration and invasion in cancer cells.
Collapse
Affiliation(s)
- Koh Nakayama
- Oxygen Biology Laboratory, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
31
|
Increasing CRTC1 function in the dentate gyrus during memory formation or reactivation increases memory strength without compromising memory quality. J Neurosci 2013; 32:17857-68. [PMID: 23223304 DOI: 10.1523/jneurosci.1419-12.2012] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Memory stabilization following encoding (synaptic consolidation) or memory reactivation (reconsolidation) requires gene expression and protein synthesis (Dudai and Eisenberg, 2004; Tronson and Taylor, 2007; Nader and Einarsson, 2010; Alberini, 2011). Although consolidation and reconsolidation may be mediated by distinct molecular mechanisms (Lee et al., 2004), disrupting the function of the transcription factor CREB impairs both processes (Kida et al., 2002; Mamiya et al., 2009). Phosphorylation of CREB at Ser133 recruits CREB binding protein (CBP)/p300 coactivators to activate transcription (Chrivia et al., 1993; Parker et al., 1996). In addition to this well known mechanism, CREB regulated transcription coactivators (CRTCs), previously called transducers of regulated CREB (TORC) activity, stimulate CREB-mediated transcription, even in the absence of CREB phosphorylation. Recently, CRTC1 has been shown to undergo activity-dependent trafficking from synapses and dendrites to the nucleus in excitatory hippocampal neurons (Ch'ng et al., 2012). Despite being a powerful and specific coactivator of CREB, the role of CRTC in memory is virtually unexplored. To examine the effects of increasing CRTC levels, we used viral vectors to locally and acutely increase CRTC1 in the dorsal hippocampus dentate gyrus region of mice before training or memory reactivation in context fear conditioning. Overexpressing CRTC1 enhanced both memory consolidation and reconsolidation; CRTC1-mediated memory facilitation was context specific (did not generalize to nontrained context) and long lasting (observed after virally expressed CRTC1 dissipated). CREB overexpression produced strikingly similar effects. Therefore, increasing CRTC1 or CREB function is sufficient to enhance the strength of new, as well as established reactivated, memories without compromising memory quality.
Collapse
|
32
|
Nekhai S, Kumari N, Dhawan S. Role of cellular iron and oxygen in the regulation of HIV-1 infection. Future Virol 2013; 8:301-311. [PMID: 23678366 DOI: 10.2217/fvl.13.6] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Despite efficient antiretroviral therapy, eradication of HIV-1 infection is challenging and requires novel biological insights and therapeutic strategies. Among other physiological and environmental factors, intracellular iron greatly affects HIV-1 replication. Higher iron stores were shown to be associated with faster progression of HIV-1 infection and to inversely correlate with the survival of HIV-1 infected patients. Iron is required for several steps in the HIV-1 life cycle, including reverse transcription, HIV-1 gene expression and capsid assembly. Here, the authors present a comprehensive review of the molecular mechanisms involved in iron- and oxygen-mediated regulation of HIV-1 replication. We also propose key intracellular pathways that may be involved in regulating HIV-1 replication, via protein kinase complexes, CDK9/cyclin T1 and CDK 2/cyclin E, protein phosphatase-1 and other host factors.
Collapse
Affiliation(s)
- Sergei Nekhai
- Center for Sickle Cell Disease, Department of Medicine, Howard University, 520 W Street, NW, Washington DC 20059, USA
| | | | | |
Collapse
|
33
|
Crowet JM, Lins L, Deshayes S, Divita G, Morris M, Brasseur R, Thomas A. Modeling of non-covalent complexes of the cell-penetrating peptide CADY and its siRNA cargo. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1828:499-509. [PMID: 23000699 DOI: 10.1016/j.bbamem.2012.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 08/09/2012] [Accepted: 09/07/2012] [Indexed: 01/06/2023]
Abstract
CADY is a cell-penetrating peptide spontaneously making non-covalent complexes with Short interfering RNAs (siRNAs) in water. Neither the structure of CADY nor that of the complexes is resolved. We have calculated and analyzed 3D models of CADY and of the non-covalent CADY-siRNA complexes in order to understand their formation and stabilization. Data from the ab initio calculations and molecular dynamics support that, in agreement with the experimental data, CADY is a polymorphic peptide partly helical. Taking into consideration the polymorphism of CADY, we calculated and compared several complexes with peptide/siRNA ratios of up to 40. Four complexes were run by using molecular dynamics. The initial binding of CADYs is essentially due to the electrostatic interactions of the arginines with siRNA phosphates. Due to a repetitive arginine motif (XLWR(K)) in CADY and to the numerous phosphate moieties in the siRNA, CADYs can adopt multiple positions at the siRNA surface leading to numerous possibilities of complexes. Nevertheless, several complex properties are common: an average of 14±1 CADYs is required to saturate a siRNA as compared to the 12±2 CADYs experimentally described. The 40 CADYs/siRNA that is the optimal ratio for vector stability always corresponds to two layers of CADYs per siRNA. When siRNA is covered by the first layer of CADYs, the peptides still bind despite the electrostatic repulsion. The peptide cage is stabilized by hydrophobic CADY-CADY contacts thanks to CADY polymorphism. The analysis demonstrates that the hydrophobicity, the presence of several positive charges and the disorder of CADY are mandatory to make stable the CADY-siRNA complexes.
Collapse
Affiliation(s)
- Jean-Marc Crowet
- Centre de Biophysique Moléculaire Numérique, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium.
| | | | | | | | | | | | | |
Collapse
|
34
|
Pourmousa M, Wong-ekkabut J, Patra M, Karttunen M. Molecular dynamic studies of transportan interacting with a DPPC lipid bilayer. J Phys Chem B 2012; 117:230-41. [PMID: 23214935 DOI: 10.1021/jp310255r] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Translocation of peptides through cellular membranes is a fundamental problem in developing antimicrobial peptides and in drug delivery. There is a class of peptides, known as cell-penetrating peptides, that are able to penetrate membranes without disrupting them. They can carry pharmacological compounds, thus a promising strategy for drug delivery. The physical mechanisms that facilitate translocation are not known. We have used large-scale molecular dynamics simulations to study the penetration of transportan across a zwitterionic dipalmitoyl-phosphatidyl-choline (DPPC) bilayer. We obtained the free energy profile for one peptide inside the bilayer and discuss the response of the bilayer to the presence of transportan. We also discuss the importance of lysine residues and speculate on the possible penetration mechanism of the peptide and propose a graded-like penetration process.
Collapse
Affiliation(s)
- Mohsen Pourmousa
- Department of Applied Mathematics, The University of Western Ontario, London, Ontario, Canada N6A 5B7
| | | | | | | |
Collapse
|
35
|
Mu J, Ostrowski RP, Soejima Y, Rolland WB, Krafft PR, Tang J, Zhang JH. Delayed hyperbaric oxygen therapy induces cell proliferation through stabilization of cAMP responsive element binding protein in the rat model of MCAo-induced ischemic brain injury. Neurobiol Dis 2012; 51:133-43. [PMID: 23146993 DOI: 10.1016/j.nbd.2012.11.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 11/01/2012] [Indexed: 01/04/2023] Open
Abstract
Treatments that could extend the therapeutic window of opportunity for stroke patients are urgently needed. Early administration of hyperbaric oxygen therapy (HBOT) has been proven neuroprotective in the middle cerebral artery occlusion (MCAo) in rodents. Our aim was to determine: 1) whether delayed HBOT after permanent MCAo (pMCAo) can still convey neuroprotection and restorative cell proliferation, and 2) whether these beneficial effects rely on HBO-induced activation of protein phosphatase-1γ (PP1-γ) leading to a decreased phosphorylation and ubiquitination of CREB and hence its stabilization. The experiments were performed in one hundred thirty-two male Sprague-Dawley rats with the body weight ranging from 240 to 270 g. Permanent MCAo was induced with the intraluminal filament occluding the right middle cerebral artery (MCA). In the first experiment, HBOT (2.5 ATA, 1h daily for 10 days) was started 48 h after pMCAo. Neurobehavioral deficits and infarct size as well as cyclic AMP response element-binding protein (CREB) expression and BrdU-DAB staining in the hippocampus and the peri-infarct region were evaluated on day 14 and day 28 post-MCAo. In the second experiment, HBOT (2.5 ATA, 1h) was started 3h after pMCAo. The effects of CREB siRNA or PP1-γ siRNA on HBO-induced infarct size alterations and target protein expression were studied. HBOT started with 48 h delay reduced infarct size, ameliorated neurobehavioral deficits and increased protein expression of CREB, resulting in increased cell proliferations in the hippocampus and peri-infarct region, on day 14 and day 28 post-MCAo. In the acute experiment pMCAo resulted in cerebral infarction and functional deterioration and reduced brain expression of PP1-γ, which led to increased phosphorylation and ubiquitination of CREB 24h after MCAo. However HBOT administered 3h after ischemia reversed these molecular events and resulted in CREB stabilization, infarct size reduction and neurobehavioral improvement. Gene silencing with CREB siRNA or PP1-γ siRNA reduced acute beneficial effects of HBO. In conclusion, delayed daily HBOT presented as potent neuroprotectant in pMCAo rats, increased CREB expression and signaling activity, and bolstered regenerative type cell proliferation in the injured brain. As shown in the acute experiment these effects of HBO were likely to be mediated by reducing ubiquitin-dependent CREB degradation owing to HBO-induced activation of PP1γ.
Collapse
Affiliation(s)
- Jun Mu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | |
Collapse
|
36
|
Fangradt M, Hahne M, Gaber T, Strehl C, Rauch R, Hoff P, Löhning M, Burmester GR, Buttgereit F. Human monocytes and macrophages differ in their mechanisms of adaptation to hypoxia. Arthritis Res Ther 2012; 14:R181. [PMID: 22870988 PMCID: PMC3580576 DOI: 10.1186/ar4011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 08/07/2012] [Indexed: 12/23/2022] Open
Abstract
Introduction Inflammatory arthritis is a progressive disease with chronic inflammation of joints, which is mainly characterized by the infiltration of immune cells and synovial hyperproliferation. Monocytes migrate towards inflamed areas and differentiate into macrophages. In inflamed tissues, much lower oxygen levels (hypoxia) are present in comparison to the peripheral blood. Hence, a metabolic adaptation process must take place. Other studies suggest that Hypoxia Inducible Factor 1-alpha (HIF-1α) may regulate this process, but the mechanism involved for human monocytes is not yet clear. To address this issue, we analyzed the expression and function of HIF-1α in monocytes and macrophages, but also considered alternative pathways involving nuclear factor of kappa light polypeptide gene enhancer in B-cells (NFκB). Methods Isolated human CD14+ monocytes were incubated under normoxia and hypoxia conditions with or without phorbol 12-myristate 13-acetate (PMA) stimulation, respectively. Nuclear and cytosolic fractions were prepared in order to detect HIF-1α and NFκB by immunoblot. For the experiments with macrophages, primary human monocytes were differentiated into human monocyte derived macrophages (hMDM) using human macrophage colony-stimulating factor (hM-CSF). The effects of normoxia and hypoxia on gene expression were compared between monocytes and hMDMs using quantitative PCR (quantitative polymerase chain reaction). Results We demonstrate, using primary human monocytes and hMDM, that the localization of transcription factor HIF-1α during the differentiation process is shifted from the cytosol (in monocytes) into the nucleus (in macrophages), apparently as an adaptation to a low oxygen environment. For this localization change, protein kinase C alpha/beta 1 (PKC-α/β1 ) plays an important role. In monocytes, it is NFκB1, and not HIF-1α, which is of central importance for the expression of hypoxia-adjusted genes. Conclusions These data demonstrate that during differentiation of monocytes into macrophages, crucial cellular adaptation mechanisms are decisively changed.
Collapse
|
37
|
Qin L, Bromberg-White JL, Qian CN. Opportunities and challenges in tumor angiogenesis research: back and forth between bench and bed. Adv Cancer Res 2012; 113:191-239. [PMID: 22429856 DOI: 10.1016/b978-0-12-394280-7.00006-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Angiogenesis is essential for tumor growth and metastasis. Many signaling pathways are involved in regulating tumor angiogenesis, with the vascular endothelial growth factor pathway being of particular interest. The recognition of the heterogeneity in tumor vasculature has led to better predictions of prognosis through differential analyses of the vasculature. However, the clinical benefits from antiangiogenic therapy are limited, because many antiangiogenic agents cannot provide long-term survival benefits, suggesting the development of drug resistance. Activation of the hypoxia and c-Met pathways, as well as other proangiogenic factors, has been shown to be responsible for such resistance. Vessel co-option could be another important mechanism. For future development, research to improve the efficacy of antiangiogenic therapy includes (a) using tumor-derived endothelial cells for drug screening; (b) developing the drugs focusing on specific tumor types; (c) developing a better preclinical model for drug study; (d) developing more accurate biomarkers for patient selection; (e) targeting the c-Met pathway or other pathways; and (f) optimizing the dose and schedule of antiangiogenic therapy. In summary, the future of antiangiogenic therapy for cancer patients depends on our efforts to develop the right drugs, select the right patients, and optimize the treatment conditions.
Collapse
Affiliation(s)
- Li Qin
- State Key Laboratory on Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, PR China
| | | | | |
Collapse
|
38
|
Velmurugan K, Balamurugan AN, Loganathan G, Ahmad A, Hering BJ, Pugazhenthi S. Antiapoptotic actions of exendin-4 against hypoxia and cytokines are augmented by CREB. Endocrinology 2012; 153:1116-28. [PMID: 22253425 DOI: 10.1210/en.2011-1895] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Islets isolated from cadaveric donor pancreas are functionally viable and can be transplanted in diabetic patients to reduce insulin requirements. This therapeutic approach is less efficient because a significant portion of functional islets is lost due to oxidative stress, inflammation, and hypoxia. Exendin-4, a glucagon-like peptide-1 receptor agonist, is known to improve islet survival through activation of the transcription factor, cAMP response element binding protein (CREB). However, isolated human islets are exposed to several stresses known to down-regulate CREB. The objective of the present study was to determine whether the cytoprotective actions of exendin-4 in human islets can be augmented by increasing the levels of CREB. Simulation of ischemia/reperfusion injury and exposure to hypoxic conditions in cultured human islets resulted in decreased CREB activation and induction of apoptosis. Islets were transduced with adenoviral CREB followed by exposure to exendin-4 as a strategy for improving their survival. This combination increased the levels of several proteins needed for β-cell survival and function, including insulin receptor substrate-2, Bcl-2, and baculoviral IAP repeat-containing 3, and suppressed the expression of proapoptotic and inflammatory genes. A combination of CREB and exendin-4 exerted enhanced antiapoptotic action in cultured islets against hypoxia and cytokines. More significantly, transplantation of human islets transduced with adenoviral CREB and treated with exendin-4 showed improved glycemic control over a 30-d period in diabetic athymic nude mice. These observations have significant implications in the therapeutic potential of exendin-4 and CREB in the islet transplantation setting as well as in preserving β-cell mass of diabetic patients.
Collapse
Affiliation(s)
- Kalpana Velmurugan
- Section of Endocrinology, Veterans Affairs Medical Center, Denver, Colorado 80220, USA
| | | | | | | | | | | |
Collapse
|
39
|
Wang YE, Pernet O, Lee B. Regulation of the nucleocytoplasmic trafficking of viral and cellular proteins by ubiquitin and small ubiquitin-related modifiers. Biol Cell 2011; 104:121-38. [PMID: 22188262 PMCID: PMC3625690 DOI: 10.1111/boc.201100105] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 11/22/2011] [Indexed: 12/29/2022]
Abstract
Nucleocytoplasmic trafficking of many cellular proteins is regulated by nuclear import/export signals as well as post-translational modifications such as covalent conjugation of ubiquitin and small ubiquitin-related modifiers (SUMOs). Ubiquitination and SUMOylation are rapid and reversible ways to modulate the intracellular localisation and function of substrate proteins. These pathways have been co-opted by some viruses, which depend on the host cell machinery to transport their proteins in and out of the nucleus. In this review, we will summarise our current knowledge on the ubiquitin/SUMO-regulated nuclear/subnuclear trafficking of cellular proteins and describe examples of viral exploitation of these pathways.
Collapse
Affiliation(s)
- Yao E Wang
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
40
|
Mu J, Krafft PR, Zhang JH. Hyperbaric oxygen therapy promotes neurogenesis: where do we stand? Med Gas Res 2011; 1:14. [PMID: 22146131 PMCID: PMC3231808 DOI: 10.1186/2045-9912-1-14] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Accepted: 06/27/2011] [Indexed: 11/10/2022] Open
Abstract
Neurogenesis in adults, initiated by injury to the central nervous system (CNS) presents an autologous repair mechanism. It has been suggested that hyperbaric oxygen therapy (HBOT) enhances neurogenesis which accordingly may improve functional outcome after CNS injury. In this present article we aim to review experimental as well as clinical studies on the subject of HBOT and neurogenesis. We demonstrate hypothetical mechanism of HBOT on cellular transcription factors including hypoxia-inducible factors (HIFs) and cAMP response element binding (CREB). We furthermore reveal the discrepancy between experimental findings and clinical trials in regards of HBOT. Further translational preclinical studies followed by improved clinical trials are needed to elucidate potential benefits of HBOT.
Collapse
Affiliation(s)
- Jun Mu
- Department of Physiology, Loma Linda University School of Medicine, 11021 Campus Street, Loma Linda, CA 92354, USA.
| | | | | |
Collapse
|
41
|
Martin SK, Diamond P, Gronthos S, Peet DJ, Zannettino ACW. The emerging role of hypoxia, HIF-1 and HIF-2 in multiple myeloma. Leukemia 2011; 25:1533-42. [PMID: 21637285 DOI: 10.1038/leu.2011.122] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hypoxia is an imbalance between oxygen supply and demand, which deprives cells or tissues of sufficient oxygen. It is well-established that hypoxia triggers adaptive responses, which contribute to short- and long-term pathologies such as inflammation, cardiovascular disease and cancer. Induced by both microenvironmental hypoxia and genetic mutations, the elevated expression of the hypoxia-inducible transcription factor-1 (HIF-1) and HIF-2 is a key feature of many human cancers and has been shown to promote cellular processes, which facilitate tumor progression. In this review, we discuss the emerging role of hypoxia and the HIFs in the pathogenesis of multiple myeloma (MM), an incurable hematological malignancy of BM PCs, which reside within the hypoxic BM microenvironment. The need for current and future therapeutic interventions to target HIF-1 and HIF-2 in myeloma will also be discussed.
Collapse
Affiliation(s)
- S K Martin
- Division of Haematology, Centre for Cancer Biology, SA Pathology, CSCR, University of Adelaide, Adelaide, South Australia
| | | | | | | | | |
Collapse
|
42
|
LIU YU, PATEL SANJAY, NIBBE ROD, MAXWELL SEAN, CHOWDHURY SALIMA, KOYUTURK MEHMET, ZHU XIAOFENG, LARKIN EMMAK, BUXBAUM SARAHG, PUNJABI NARESHM, GHARIB SINAA, REDLINE SUSAN, CHANCE MARKR. Systems biology analyses of gene expression and genome wide association study data in obstructive sleep apnea. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2011:14-25. [PMID: 21121029 PMCID: PMC4465214 DOI: 10.1142/9789814335058_0003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The precise molecular etiology of obstructive sleep apnea (OSA) is unknown; however recent research indicates that several interconnected aberrant pathways and molecular abnormalities are contributors to OSA. Identifying the genes and pathways associated with OSA can help to expand our understanding of the risk factors for the disease as well as provide new avenues for potential treatment. Towards these goals, we have integrated relevant high dimensional data from various sources, such as genome-wide expression data (microarray), protein-protein interaction (PPI) data and results from genome-wide association studies (GWAS) in order to define sub-network elements that connect some of the known pathways related to the disease as well as define novel regulatory modules related to OSA. Two distinct approaches are applied to identify sub-networks significantly associated with OSA. In the first case we used a biased approach based on sixty genes/proteins with known associations with sleep disorders and/or metabolic disease to seed a search using commercial software to discover networks associated with disease followed by information theoretic (mutual information) scoring of the sub-networks. In the second case we used an unbiased approach and generated an interactome constructed from publicly available gene expression profiles and PPI databases, followed by scoring of the network with p-values from GWAS data derived from OSA patients to uncover sub-networks significant for the disease phenotype. A comparison of the approaches reveals a number of proteins that have been previously known to be associated with OSA or sleep. In addition, our results indicate a novel association of Phosphoinositide 3-kinase, the STAT family of proteins and its related pathways with OSA.
Collapse
Affiliation(s)
- YU LIU
- Center for Proteomics & Bioinformatics, Case Western Reserve University (CWRU), Cleveland, Ohio, 44106, USA
| | - SANJAY PATEL
- Division of Pulmonary, Critical Care and Sleep Medicine, CWRU, Cleveland, Ohio, 44106, USA
| | - ROD NIBBE
- Center for Proteomics & Bioinformatics, CWRU, Cleveland, Ohio, 44106, USA
| | - SEAN MAXWELL
- Center for Proteomics & Bioinformatics, CWRU, Cleveland, Ohio, 44106, USA
| | - SALIM A. CHOWDHURY
- Department of Electrical Engineering & Computer Science, CWRU, Cleveland, Ohio, 44106, USA
| | - MEHMET KOYUTURK
- Department of Electrical Engineering & Computer Science, CWRU, Cleveland, Ohio, 44106, USA
| | - XIAOFENG ZHU
- Department of Epidemiology and Biostatistics, CWRU, Cleveland, Ohio, 44106, USA
| | - EMMA K. LARKIN
- Division of Allergy, Pulmonary and Critical Care, Vanderbilt University Medical Center, 1215 21st Ave S., Nashville, Tennessee, 37232, USA
| | - SARAH G BUXBAUM
- Jackson Heart Study, Jackson State University, Jackson, MS 39213, USA
| | - NARESH M. PUNJABI
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - SINA A. GHARIB
- Center for Lung Biology, Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA 98109, USA
| | - SUSAN REDLINE
- Department of Medicine, CWRU, Cleveland, Ohio, 44106, and Depart of Medicine, Brigham & Women’s Hospital and Beth Israel Deaconess Medical School, Harvard Medical School, Boston, MA, 02115
| | - MARK R. CHANCE
- Center for Proteomics & Bioinformatics, Department of Genetics, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| |
Collapse
|
43
|
Sakamoto K, Karelina K, Obrietan K. CREB: a multifaceted regulator of neuronal plasticity and protection. J Neurochem 2010; 116:1-9. [PMID: 21044077 DOI: 10.1111/j.1471-4159.2010.07080.x] [Citation(s) in RCA: 386] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Since its initial characterization over 20 years ago, there has been intense and unwavering interest in understanding the role of the transcription factor cAMP-responsive element binding protein (CREB) in nervous system physiology. Through an array of experimental approaches and model systems, researchers have begun to unravel the complex and multifaceted role of this transcription factor in such diverse processes as neurodevelopment, synaptic plasticity, and neuroprotection. Here we discuss current insights into the molecular mechanisms by which CREB couples synaptic activity to long-term changes in neuronal plasticity, which is thought to underlie learning and memory. We also discuss work showing that CREB is a critical component of the neuroprotective transcriptional network, and data indicating that CREB dysregulation contributes to an array of neuropathological conditions.
Collapse
Affiliation(s)
- Kensuke Sakamoto
- Department of Neuroscience, Ohio State University, Columbus, Ohio 43210, USA
| | | | | |
Collapse
|
44
|
Liu L, Zhang H, Sun L, Gao Y, Jin H, Liang S, Wang Y, Dong M, Shi Y, Li Z, Fan D. ERK/MAPK activation involves hypoxia-induced MGr1-Ag/37LRP expression and contributes to apoptosis resistance in gastric cancer. Int J Cancer 2010; 127:820-829. [PMID: 19998339 DOI: 10.1002/ijc.25098] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We previously demonstrated that hypoxia increased the hypoxia-inducible factor (HIF-1)-dependent MGr1-Ag/37LRP expression, which enhanced adhesion of gastric cancer cells to laminin, inhibited drug-induced apoptosis and caused cell adhesion-mediated drug resistance (CAM-DR). Here, we investigated the role of extracellular-regulated kinase (ERK) 1/2 in the signaling mechanisms underlying these events. We found that hypoxia activated ERK activity in vitro and in vivo. Overexpression of mitogen-activated protein kinase (MAPK) kinase (MEK), which preferentially activated ERK, mimics, in a nonadditive way, hypoxia-induced activity of MGr1-Ag/37LRP promoter and expression of MGr1-Ag/37LRP. Furthermore, U0126, the MEK inhibitor, inhibited hypoxia- and MEK-induced MGr1-Ag/37LRP promoter activity in a dose-dependent manner. MEK inhibition also reversed hypoxia- and MEK-induced HIF-1 protein and its activity in a dose-dependent manner. We also investigated reactive oxygen species signaling this response. Exogenous addition of H(2)O(2) was sufficient to activate ERK in a dose-dependent profile. Reactive oxygen species scavengers of H(2)O(2) significantly inhibited hypoxia-induced ERK or HIF-1 activation and sequential expression of MGr1-Ag/37LRP. We also investigated the signaling in hypoxia-induced cell adhesion and apoptosis induced by vincristine. Hypoxia significantly enhanced adhesion of SGC7901 cells to laminin in a time-dependent manner, which might be inhibited by the MEK inhibitor U0126 and MGr1-Ag/37LRP siRNA. Consistent with results of adhesion assay, hypoxia-resistant apoptosis might be reversed by U0126 in a dose-dependent manner. Our results suggest that hypoxia-elicited MGr1-Ag/37LRP expression activated by HIF-1 depends on ERK activation. These events are dependent of reactive oxygen intermediates.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Apoptosis
- Blotting, Western
- Butadienes/pharmacology
- Cell Adhesion
- Cell Movement
- Dose-Response Relationship, Drug
- Enzyme Inhibitors/pharmacology
- Enzyme-Linked Immunosorbent Assay
- Humans
- Hydrogen Peroxide/pharmacology
- Hypoxia/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Mice
- Mitogen-Activated Protein Kinase 1/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 1/physiology
- Mitogen-Activated Protein Kinase 3/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 3/physiology
- Nitriles/pharmacology
- Oxidants/pharmacology
- Phosphorylation
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/pharmacology
- Reactive Oxygen Species/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Stomach Neoplasms/metabolism
- Stomach Neoplasms/pathology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Lili Liu
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ozgen N, Lau DH, Shlapakova IN, Sherman W, Feinmark SJ, Danilo P, Rosen MR. Determinants of CREB degradation and KChIP2 gene transcription in cardiac memory. Heart Rhythm 2010; 7:964-70. [PMID: 20346417 DOI: 10.1016/j.hrthm.2010.03.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Accepted: 03/19/2010] [Indexed: 01/12/2023]
Abstract
BACKGROUND Left ventricular pacing (LVP) to induce cardiac memory (CM) in dogs results in a decreased transient outward K current (I(to)) and reduced mRNA and protein of the I(to) channel accessory subunit, KChIP2. The KChIP2 decrease is attributed to a decrease in its transcription factor, cyclic adenosine monophosphate response element binding protein (CREB). OBJECTIVE This study sought to determine the mechanisms responsible for the CREB decrease that is initiated by LVP. METHODS CM was quantified as T-wave vector displacement in 18 LVP dogs. In 5 dogs, angiotensin II receptor blocker, saralasin, was infused before and during pacing. In 3 dogs, proteasomal inhibitor, lactacystin, was injected into the left anterior descending artery before LVP. Epicardial biopsy samples were taken before and after LVP. Neonatal rat cardiomyocytes (NRCM) were incubated with H(2)O(2) (50 micromol/l) for 1 hour with or without lactacystin. RESULTS LVP significantly displaced the T-wave vector and was associated with increased lipid peroxidation and increased tissue angiotensin II levels. Saralasin prevented T-vector displacement and lipid peroxidation. CREB was significantly decreased after 2 hours of LVP and was comparably decreased in H(2)O(2)-treated NRCM. Lactacystin inhibited the CREB decrease in LVP dogs and H(2)O(2)-treated NRCM. LVP and H(2)O(2) both induced CREB ubiquitination, and the H(2)O(2)-induced CREB decrease was prevented by knocking down ubiquitin. CONCLUSION LVP initiates myocardial angiotensin II production and reactive oxygen species synthesis, leading to CREB ubiquitination and its proteasomal degradation. This sequence of events would explain the pacing-induced reduction in KChIP2, and contribute to altered repolarization and the T-wave changes of cardiac memory.
Collapse
Affiliation(s)
- Nazira Ozgen
- Department of Pharmacology, Columbia University, New York, New York 10032, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
SUMO and ubiquitin modifications during steroid hormone synthesis and function. Biochem Soc Trans 2010; 38:54-9. [DOI: 10.1042/bst0380054] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Steroid hormones control many aspects of animal physiology and behaviour. They are highly regulated, among other mechanisms, by post-translational modifications of the transcription factors involved in their synthesis and response. In the present review, we will focus on the influence of SUMO (small ubiquitin-related modifier) and ubiquitin modifications on the function of transcription factors involved in adrenal cortex formation, steroidogenesis and the hormonal response.
Collapse
|
47
|
Ozgen N, Guo J, Gertsberg Z, Danilo P, Rosen MR, Steinberg SF. Reactive oxygen species decrease cAMP response element binding protein expression in cardiomyocytes via a protein kinase D1-dependent mechanism that does not require Ser133 phosphorylation. Mol Pharmacol 2009; 76:896-902. [PMID: 19620255 PMCID: PMC2769048 DOI: 10.1124/mol.109.056473] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Accepted: 07/17/2009] [Indexed: 12/21/2022] Open
Abstract
Reactive oxygen species (ROS) exert pleiotropic effects on a wide array of signaling proteins that regulate cellular growth and apoptosis. This study shows that long-term treatment with a low concentration of H2O2 leads to the activation of signaling pathways involving extracellular signal-regulated kinase, ribosomal protein S6 kinase, and protein kinase D (PKD) that increase cAMP binding response element protein (CREB) phosphorylation at Ser(133) in cardiomyocytes. Although CREB-Ser(133) phosphorylation typically mediates cAMP-dependent increases in CREB target gene expression, the H2O2-dependent increase in CREB-Ser(133) phosphorylation is accompanied by a decrease in CREB protein abundance and no change in Cre-luciferase reporter activity. Mutagenesis studies indicate that H2O2 decreases CREB protein abundance via a mechanism that does not require CREB-Ser(133) phosphorylation. Rather, the H2O2-dependent decrease in CREB protein is prevented by the proteasome inhibitor lactacystin, by inhibitors of mitogen-activated protein kinase kinase or protein kinase C activity, or by adenoviral-mediated delivery of a small interfering RNA that decreases PKD1 expression. A PKD1-dependent mechanism that links oxidative stress to decreased CREB protein abundance is predicted to contribute to the pathogenesis of heart failure by influencing cardiac growth and apoptosis responses.
Collapse
Affiliation(s)
- Nazira Ozgen
- Center for Molecular Therapeutics, Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | |
Collapse
|
48
|
Charles S, Ammosova T, Cardenas J, Foster A, Rotimi J, Jerebtsova M, Ayodeji AA, Niu X, Ray PE, Gordeuk VR, Kashanchi F, Nekhai S. Regulation of HIV-1 transcription at 3% versus 21% oxygen concentration. J Cell Physiol 2009; 221:469-79. [PMID: 19626680 DOI: 10.1002/jcp.21882] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
HIV transcription is induced by the HIV-1 Tat protein, in concert with cellular co-factors including CDK9, CDK2, NF-kappaB, and others. The cells of most of the body's organs are exposed to approximately 3-6% oxygen, but most in vitro studies of HIV replication are conducted at 21% oxygen. We hypothesized that activities of host cell factors involved in HIV-1 replication may differ at 3% versus 21% O(2), and that such differences may affect HIV-1 replication. Here we show that Tat-induced HIV-1 transcription was reduced at 3% O(2) compared to 21% O(2). HIV-1 replication was also reduced in acutely or chronically infected cells cultured at 3% O(2) compared to 21% O(2). This reduction was not due the decreased cell growth or increased cellular toxicity and also not due to the induction of hypoxic response. At 3% O(2), the activity of CDK9/cyclin T1 was inhibited and Sp1 activity was reduced, whereas the activity of other host cell factors such as CDK2 or NF-kappaB was not affected. CDK9-specific inhibitor ARC was much less efficient at 3% compared to 21% O(2) and also expression of CDK9/cyclin T1-dependent IkappaB inhibitor alpha was repressed. Our results suggest that lower HIV-1 transcription at 3% O(2) compared to 21% O(2) may be mediated by lower activity of CDK9/cyclin T1 and Sp1 at 3% O(2) and that additional host cell factors such as CDK2 and NF-kappaB might be major regulators of HIV-1 transcription at low O(2) concentrations.
Collapse
Affiliation(s)
- Sharroya Charles
- Center for Sickle Cell Disease, Howard University, Washington, DC 20001, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Alvarez-Castelao B, Martín-Guerrero I, García-Orad Á, Castaño JG. Cytomegalovirus promoter up-regulation is the major cause of increased protein levels of unstable reporter proteins after treatment of living cells with proteasome inhibitors. J Biol Chem 2009; 284:28253-28262. [PMID: 19679666 DOI: 10.1074/jbc.m109.004101] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Fluorescent unstable proteins obtained by the fusion of a fluorescent protein coding sequence with specific amino acid sequences that promote its fast degradation have become popular to gauge the activity of the ubiquitin/proteasome system in living cells. The steady-state levels of expression of these unstable proteins is low in agreement with their short half-lives, and they accumulate in the cell upon treatment with proteasome inhibitors. We show here that this accumulation is mainly due to transcriptional up-regulation of the cytomegalovirus promoter by proteasome inhibitors and mediated, at least in part, by AP1 transactivation. These simple facts put under quarantine conclusions reached about the activity of the ubiquitin/proteasome pathway in animal cells in culture or in transgenic mice, where popular cytomegalovirus-driven constructs are used, as transcriptional regulation of the expression of the reporter protein construct and not degradation of the unstable protein by the ubiquitin/proteasome system may contribute significantly to the interpretation of the results observed.
Collapse
Affiliation(s)
- Beatriz Alvarez-Castelao
- Departamento de Bioquímica, Instituto de Investigaciones Biomédicas "Alberto Sols," Universidad Autónoma de Madrid y Consejo Superior de Investigaciones Científicas (UAM-CSIC) y Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Facultad de Medicina UAM, 28029 Madrid, Spain
| | - Idoia Martín-Guerrero
- Departamento de Genética, Antropología Física y Fisiología Animal, Facultad de Medicina, Universidad del País Vasco (UPV/EHU), 48940 Leioa, Spain
| | - África García-Orad
- Departamento de Genética, Antropología Física y Fisiología Animal, Facultad de Medicina, Universidad del País Vasco (UPV/EHU), 48940 Leioa, Spain
| | - José G Castaño
- Departamento de Bioquímica, Instituto de Investigaciones Biomédicas "Alberto Sols," Universidad Autónoma de Madrid y Consejo Superior de Investigaciones Científicas (UAM-CSIC) y Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Facultad de Medicina UAM, 28029 Madrid, Spain.
| |
Collapse
|
50
|
Torii S, Okamura N, Suzuki Y, Ishizawa T, Yasumoto KI, Sogawa K. Cyclic AMP represses the hypoxic induction of hypoxia-inducible factors in PC12 cells. J Biochem 2009; 146:839-44. [PMID: 19671538 DOI: 10.1093/jb/mvp129] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Hypoxia-inducible factor 1 (HIF-1) is a master regulator for hypoxic activation of genes for angiogenesis, hormone synthesis, glycolysis and cell survival. In addition to hypoxic stimulus, various effectors and reagents were reported to affect HIF-1 activity. Here, we show that cyclic AMP (cAMP) down-regulates the HIF-1 activity in pheochromocytoma PC12 cells but not in Hep3B and HeLa cells. Hypoxia response element-dependent reporter activity was decreased by the addition of dibutyryl cAMP. Expression of protein kinase A (PKA) catalytic alpha-subunits repressed the HIF-1 activity. HIF-1alpha and HLF (HIF-2alpha or EPAS1) protein levels were decreased by the treatment with dibutyryl cAMP. Although CREB was served as a negative factor for the HIF-1 activity, it may not be a major PKA target in the cAMP-dependent HIF-alpha repression pathway. Induction of hypoxia responsive genes was suppressed by dibutyryl cAMP. Our results provide additional insight into a regulatory mechanism of hypoxic response.
Collapse
Affiliation(s)
- Satoru Torii
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Japan
| | | | | | | | | | | |
Collapse
|