201
|
Wang Y, Hersheson J, Lopez D, Hammer M, Liu Y, Lee KH, Pinto V, Seinfeld J, Wiethoff S, Sun J, Amouri R, Hentati F, Baudry N, Tran J, Singleton AB, Coutelier M, Brice A, Stevanin G, Durr A, Bi X, Houlden H, Baudry M. Defects in the CAPN1 Gene Result in Alterations in Cerebellar Development and Cerebellar Ataxia in Mice and Humans. Cell Rep 2016; 16:79-91. [PMID: 27320912 DOI: 10.1016/j.celrep.2016.05.044] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 04/14/2016] [Accepted: 05/10/2016] [Indexed: 11/16/2022] Open
Abstract
A CAPN1 missense mutation in Parson Russell Terrier dogs is associated with spinocerebellar ataxia. We now report that homozygous or heterozygous CAPN1-null mutations in humans result in cerebellar ataxia and limb spasticity in four independent pedigrees. Calpain-1 knockout (KO) mice also exhibit a mild form of ataxia due to abnormal cerebellar development, including enhanced neuronal apoptosis, decreased number of cerebellar granule cells, and altered synaptic transmission. Enhanced apoptosis is due to absence of calpain-1-mediated cleavage of PH domain and leucine-rich repeat protein phosphatase 1 (PHLPP1), which results in inhibition of the Akt pro-survival pathway in developing granule cells. Injection of neonatal mice with the indirect Akt activator, bisperoxovanadium, or crossing calpain-1 KO mice with PHLPP1 KO mice prevented increased postnatal cerebellar granule cell apoptosis and restored granule cell density and motor coordination in adult mice. Thus, mutations in CAPN1 are an additional cause of ataxia in mammals, including humans.
Collapse
Affiliation(s)
- Yubin Wang
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Joshua Hersheson
- The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Dulce Lopez
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Monia Hammer
- Department of Molecular Neurobiology and Neuropathology, National Institute of Neurology, La Rabta, Tunis 1007, Tunisia; Laboratory of Neurogenetics, National Institutes of Health, Bethesda 20892, MD, USA
| | - Yan Liu
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Ka-Hung Lee
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Vanessa Pinto
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Jeff Seinfeld
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Sarah Wiethoff
- The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK; Center for Neurology and Hertie Institute for Clinical Brain Research, Eberhard-Karls-University, 72076 Tübingen, Germany
| | - Jiandong Sun
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Rim Amouri
- Department of Molecular Neurobiology and Neuropathology, National Institute of Neurology, La Rabta, Tunis 1007, Tunisia
| | - Faycal Hentati
- Department of Molecular Neurobiology and Neuropathology, National Institute of Neurology, La Rabta, Tunis 1007, Tunisia
| | - Neema Baudry
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Jennifer Tran
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institutes of Health, Bethesda 20892, MD, USA
| | - Marie Coutelier
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, Université Pierre et Marie Curie Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, 75013 Paris, France; Laboratory of Human Molecular Genetics, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium; Ecole Pratique des Hautes Etudes (EPHE), Paris Sciences et Lettres (PSL) Research University, 75013 Paris, France
| | - Alexis Brice
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, Université Pierre et Marie Curie Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, 75013 Paris, France; Centre de Référence de Neurogénétique, Hôpital de la Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris, 75013 Paris, France
| | - Giovanni Stevanin
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, Université Pierre et Marie Curie Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, 75013 Paris, France; Ecole Pratique des Hautes Etudes (EPHE), Paris Sciences et Lettres (PSL) Research University, 75013 Paris, France; Centre de Référence de Neurogénétique, Hôpital de la Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris, 75013 Paris, France
| | - Alexandra Durr
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, Université Pierre et Marie Curie Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, 75013 Paris, France; Centre de Référence de Neurogénétique, Hôpital de la Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris, 75013 Paris, France
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Henry Houlden
- The National Hospital for Neurology and Neurosurgery and UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.
| | - Michel Baudry
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA.
| |
Collapse
|
202
|
Lui GYL, Kovacevic Z, Richardson V, Merlot AM, Kalinowski DS, Richardson DR. Targeting cancer by binding iron: Dissecting cellular signaling pathways. Oncotarget 2016; 6:18748-79. [PMID: 26125440 PMCID: PMC4662454 DOI: 10.18632/oncotarget.4349] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/12/2015] [Indexed: 12/30/2022] Open
Abstract
Newer and more potent therapies are urgently needed to effectively treat advanced cancers that have developed resistance and metastasized. One such strategy is to target cancer cell iron metabolism, which is altered compared to normal cells and may facilitate their rapid proliferation. This is supported by studies reporting the anti-neoplastic activities of the clinically available iron chelators, desferrioxamine and deferasirox. More recently, ligands of the di-2-pyridylketone thiosemicarbazone (DpT) class have demonstrated potent and selective anti-proliferative activity across multiple cancer-types in vivo, fueling studies aimed at dissecting their molecular mechanisms of action. In the past five years alone, significant advances have been made in understanding how chelators not only modulate cellular iron metabolism, but also multiple signaling pathways implicated in tumor progression and metastasis. Herein, we discuss recent research on the targeting of iron in cancer cells, with a focus on the novel and potent DpT ligands. Several key studies have revealed that iron chelation can target the AKT, ERK, JNK, p38, STAT3, TGF-β, Wnt and autophagic pathways to subsequently inhibit cellular proliferation, the epithelial-mesenchymal transition (EMT) and metastasis. These developments emphasize that these novel therapies could be utilized clinically to effectively target cancer.
Collapse
Affiliation(s)
- Goldie Y L Lui
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Zaklina Kovacevic
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Vera Richardson
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Angelica M Merlot
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Danuta S Kalinowski
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Des R Richardson
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
203
|
Akt1-mediated Gata3 phosphorylation controls the repression of IFNγ in memory-type Th2 cells. Nat Commun 2016; 7:11289. [PMID: 27053161 PMCID: PMC4829694 DOI: 10.1038/ncomms11289] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 03/09/2016] [Indexed: 12/21/2022] Open
Abstract
Th2 cells produce Th2 cytokines such as IL-4, IL-5 and IL-13, but repress Th1 cytokine IFNγ. Recent studies have revealed various distinct memory-type Th2 cell subsets, one of which produces a substantial amount of IFNγ in addition to Th2 cytokines, however it remains unclear precisely how these Th2 cells produce IFNγ. We herein show that phosphorylation of Gata3 at Ser308, Thr315 and Ser316 induces dissociation of a histone deacetylase Hdac2 from the Gata3/Chd4 repressive complex in Th2 cells. We also identify Akt1 as a Gata3-phosphorylating kinase, and the activation of Akt1 induces derepression of Tbx21 and Ifng expression in Th2 cells. Moreover, T-bet-dependent IFNγ expression in IFNγ-producing memory Th2 cells appears to be controlled by the phosphorylation status of Gata3 in human and murine systems. Thus, this study highlights the molecular basis for posttranslational modifications of Gata3 that control the regulation of IFNγ expression in memory Th2 cells.
Collapse
|
204
|
Sen T, Sen N. Treatment with an activator of hypoxia-inducible factor 1, DMOG provides neuroprotection after traumatic brain injury. Neuropharmacology 2016; 107:79-88. [PMID: 26970014 DOI: 10.1016/j.neuropharm.2016.03.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 01/25/2016] [Accepted: 03/04/2016] [Indexed: 12/26/2022]
Abstract
Traumatic brain injury (TBI) is one of the major cause of morbidity and mortality and it affects more than 1.7 million people in the USA. A couple of regenerative pathways including activation of hypoxia-inducible transcription factor 1 alpha (HIF-1α) are initiated to reduce cellular damage following TBI; however endogenous activation of these pathways is not enough to provide neuroprotection after TBI. Thus we aimed to see whether sustained activation of HIF-1α can provide neuroprotection and neurorepair following TBI. We found that chronic treatment with dimethyloxaloylglycine (DMOG) markedly increases the expression level of HIF-1α and mRNA levels of its downstream proteins such as Vascular endothelial growth factor (VEGF), Phosphoinositide-dependent kinase-1 and 4 (PDK1, PDK4) and Erythropoietin (EPO). Treatment of DMOG activates a major cell survival protein kinase Akt and reduces both cell death and lesion volume following TBI. Moreover, administration of DMOG augments cluster of differentiation 31 (CD31) staining in pericontusional cortex after TBI, which suggests that DMOG stimulates angiogenesis after TBI. Treatment with DMOG also improves both memory and motor functions after TBI. Taken together our results suggest that sustained activation of HIF-1α provides significant neuroprotection following TBI.
Collapse
Affiliation(s)
- Tanusree Sen
- Department of Neuroscience and Regenerative Medicine, Augusta University, United States; Department of Veterinary Biosciences & Diagnostic Imaging, College of Veterinary Medicine, The University of Georgia, United States
| | - Nilkantha Sen
- Department of Neuroscience and Regenerative Medicine, Augusta University, United States.
| |
Collapse
|
205
|
OUYANG QIANWEN, ZHANG LONG, JIANG YIZHOU, NI XIAOJIAN, CHEN SHENG, YE FUGUI, DU YIQUN, HUANG LIANG, DING PEIPEI, WANG NA, YANG CHAOQUN, HUANG TIANBAO, SUN YUJING, LI SHAN, XIA YUN, HU WEIGUO, LUO RONGCHENG, SHAO ZHIMING. The membrane complement regulatory protein CD59 promotes tumor growth and predicts poor prognosis in breast cancer. Int J Oncol 2016; 48:2015-24. [DOI: 10.3892/ijo.2016.3408] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/01/2016] [Indexed: 11/06/2022] Open
|
206
|
Xu Y, Xu Y, Luan H, Jiang Y, Tian X, Zhang S. Cardioprotection against experimental myocardial ischemic injury using cornin. Braz J Med Biol Res 2016; 49:e5039. [PMID: 26871971 PMCID: PMC4742973 DOI: 10.1590/1414-431x20155039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/13/2015] [Indexed: 02/03/2023] Open
Abstract
Phosphorylated-cyclic adenosine monophosphate response element-binding protein (Phospho-CREB) has an important role in the pathogenesis of myocardial ischemia. We isolated the iridoid glycoside cornin from the fruit of Verbena officinalis L, investigated its effects against myocardial ischemia and reperfusion (I/R) injury in vivo, and elucidated its potential mechanism in vitro. Effects of cornin on cell viability, as well as expression of phospho-CREB and phospho-Akt in hypoxic H9c2 cells in vitro, and myocardial I/R injury in vivo, were investigated. Cornin attenuated hypoxia-induced cytotoxicity significantly in H9c2 cells in a concentration-dependent manner. Treatment of H9c2 cells with cornin (10 µM) blocked the reduction of expression of phospho-CREB and phospho-Akt in a hypoxic condition. Treatment of rats with cornin (30 mg/kg, iv) protected them from myocardial I/R injury as indicated by a decrease in infarct volume, improvement in hemodynamics, and reduction of severity of myocardial damage. Cornin treatment also attenuated the reduction of expression of phospho-CREB and phospho-Akt in ischemic myocardial tissue. These data suggest that cornin exerts protective effects due to an increase in expression of phospho-CREB and phospho-Akt.
Collapse
Affiliation(s)
- Y Xu
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China
| | - Y Xu
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China
| | - H Luan
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China
| | - Y Jiang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China
| | - X Tian
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China
| | - S Zhang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China
| |
Collapse
|
207
|
Rago V, Romeo F, Giordano F, Ferraro A, Carpino A. Identification of the G protein-coupled estrogen receptor (GPER) in human prostate: expression site of the estrogen receptor in the benign and neoplastic gland. Andrology 2015; 4:121-7. [PMID: 26714890 DOI: 10.1111/andr.12131] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 10/02/2015] [Accepted: 10/19/2015] [Indexed: 12/11/2022]
Abstract
Estrogens are involved in growth, differentiation and pathogenesis of human prostate through the mediation of the classical estrogen receptors ERα and ERβ. The G protein-coupled estrogen receptor (GPER) is a 'novel' mediator of estrogen signaling which has been recently recognized in some human reproductive tissues, but its expression in the prostate gland is still unknown. Here, we investigated GPER in benign (from 5 patients) and neoplastic prostatic tissues (from 50 patients) by immunohistochemical analysis and Western blotting. Normal areas of benign prostates revealed a strong GPER immunoreactivity in the basal epithelial cells while luminal epithelial cells were unreactive and stromal cells were weakly immunostained. GPER was also immunolocalized in adenocarcinoma samples but the immunoreactivity of tumoral areas decreased from Gleason pattern 2 to Gleason pattern 4. Furthermore, a strong GPER immunostaining was also revealed in cells of pre-neoplastic lesions (high-grade prostatic intra-epithelial neoplasia). Western blot analysis of benign and tumor protein extracts showed the presence of a ~42 kDa band, consistent with the GPER molecular weight. An increase in both pAkt and p cAMP-response-binding protein (pCREB) levels was also observed in poorly differentiated PCa samples. Finally, this work identified GPER in the epithelial basal cells of benign human prostate, with a different localization with respect to the classical estrogen receptors. Furthermore, the expression of GPER in prostatic adenocarcinoma cells was also observed but with a modulation of the immunoreactivity according to tumor cell arrangements.
Collapse
Affiliation(s)
- V Rago
- Department of Pharmacy, Health Science and Nutrition, University of Calabria, Cosenza, Italy
| | - F Romeo
- Pathologic Anatomy Unit, Annunziata Hospital, Cosenza, Italy
| | - F Giordano
- Department of Pharmacy, Health Science and Nutrition, University of Calabria, Cosenza, Italy
| | - A Ferraro
- Pathologic Anatomy Unit, Annunziata Hospital, Cosenza, Italy
| | - A Carpino
- Department of Pharmacy, Health Science and Nutrition, University of Calabria, Cosenza, Italy
| |
Collapse
|
208
|
Avolio E, Gianfranceschi G, Cesselli D, Caragnano A, Athanasakis E, Katare R, Meloni M, Palma A, Barchiesi A, Vascotto C, Toffoletto B, Mazzega E, Finato N, Aresu G, Livi U, Emanueli C, Scoles G, Beltrami CA, Madeddu P, Beltrami AP. Ex vivo molecular rejuvenation improves the therapeutic activity of senescent human cardiac stem cells in a mouse model of myocardial infarction. Stem Cells 2015; 32:2373-85. [PMID: 24801508 DOI: 10.1002/stem.1728] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 04/05/2014] [Accepted: 04/17/2014] [Indexed: 12/12/2022]
Abstract
Cardiac stem cells (CSC) from explanted decompensated hearts (E-CSC) are, with respect to those obtained from healthy donors (D-CSC), senescent and functionally impaired. We aimed to identify alterations in signaling pathways that are associated with CSC senescence. Additionally, we investigated if pharmacological modulation of altered pathways can reduce CSC senescence in vitro and enhance their reparative ability in vivo. Measurement of secreted factors showed that E-CSC release larger amounts of proinflammatory cytokine IL1β compared with D-CSC. Using blocking antibodies, we verified that IL1β hampers the paracrine protective action of E-CSC on cardiomyocyte viability. IL1β acts intracranially inducing IKKβ signaling, a mechanism that via nuclear factor-κB upregulates the expression of IL1β itself. Moreover, E-CSC show reduced levels of AMP protein kinase (AMPK) activating phosphorylation. This latter event, together with enhanced IKKβ signaling, increases TORC1 activity, thereby impairing the autophagic flux and inhibiting the phosphorylation of Akt and cAMP response element-binding protein. The combined use of rapamycin and resveratrol enhanced AMPK, thereby restoring downstream signaling and reducing IL1β secretion. These molecular corrections reduced E-CSC senescence, re-establishing their protective activity on cardiomyocytes. Moreover ex vivo treatment with rapamycin and resveratrol improved E-CSC capacity to induce cardiac repair upon injection in the mouse infarcted heart, leading to reduced cardiomyocyte senescence and apoptosis and increased abundance of endogenous c-Kit(+) CSC in the peri-infarct area. Molecular rejuvenation of patient-derived CSC by short pharmacologic conditioning boosts their in vivo reparative abilities. This approach might prove useful for refinement of CSC-based therapies.
Collapse
Affiliation(s)
- Elisa Avolio
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
209
|
Yi JH, Beak SJ, Lee S, Jung JW, Kim BC, Ryu JH, Kim DH. Danggui-Jakyak-San enhances hippocampal long-term potentiation through the ERK/CREB/BDNF cascade. JOURNAL OF ETHNOPHARMACOLOGY 2015; 175:481-489. [PMID: 26453932 DOI: 10.1016/j.jep.2015.10.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 07/14/2015] [Accepted: 10/04/2015] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Danggui-Jakyak-San (DJS), a traditional herbal prescription, has long been used to treat gerontological disorders due to insufficient blood supply. AIM OF THE STUDY Previously, we reported that DJS increased hippocampal neurogenesis and enhanced learning and memory. However, the precise mechanism of DJS and its effects on learning and memory are still not well understood. In this study, we investigated the effect of DJS on hippocampal long-term potentiation (LTP), a cellular mechanism thought to underlie learning and memory. MATERIALS AND METHODS To understand the effect of DJS on LTP, we used acute mouse hippocampal slices and delivered one train of high frequency stimulation (100 Hz, 100 pulses). Western blots were used to analyze the changes in protein levels induced by DJS. Morris water maze test was used to evaluate the effect of DJS on spatial long-term memory. RESULTS DJS enhanced LTP in the Schaffer-collateral pathway of the hippocampus in a concentration-dependent manner. Extracellular signal-regulated kinase 1/2 (ERK1/2) and cAMP response element-binding protein (CREB) were activated by DJS. Moreover, brain-derived neurotropic factor (BDNF) was also increased by DJS. Blockade of ERK1/2 activation with PD198306 blocked the DJS-induced activation of the ERK1/2/CREB/BDNF cascade and LTP enhancement. In vivo, DJS improved spatial long-term memory and upregulated the hippocampal CREB/BDNF cascade. CONCLUSION These results suggest that DJS enhances hippocampal LTP and spatial memory through the ERK/CREB/BDNF cascade.
Collapse
Affiliation(s)
- Jee Hyun Yi
- School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, Bristol, UK
| | - Soo Ji Beak
- Chonnam-Bristol Frontier Laboratory, Biomedical Research Institute, Chonnam National University Hospital, Jebong-ro, Gwangju 501-757, Republic of Korea
| | - Seungheon Lee
- Department of Aquatic Biomedical Sciences, School of Marine Biomedical Science, College of Ocean Science, Jeju National University, Jeju 690-756, Republic of Korea
| | - Ji Wook Jung
- Department of Herbal Medicinal Pharmacology, College of Herbal Bio-industry, Daegu Haany University, Kyungsan, Republic of Korea
| | - Byeong C Kim
- Chonnam-Bristol Frontier Laboratory, Biomedical Research Institute, Chonnam National University Hospital, Jebong-ro, Gwangju 501-757, Republic of Korea
| | - Jong Hoon Ryu
- Department of Life and Nanopharmaceutical Sciences and,College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea; Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea.
| | - Dong Hyun Kim
- Department of Medicinal Biotechnology, College of Natural Resources and Life Science, Dong-A University, Busan, Republic of Korea.
| |
Collapse
|
210
|
Yang SF, Lee WJ, Tan P, Tang CH, Hsiao M, Hsieh FK, Chien MH. Upregulation of miR-328 and inhibition of CREB-DNA-binding activity are critical for resveratrol-mediated suppression of matrix metalloproteinase-2 and subsequent metastatic ability in human osteosarcomas. Oncotarget 2015; 6:2736-53. [PMID: 25605016 PMCID: PMC4413614 DOI: 10.18632/oncotarget.3088] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 12/25/2014] [Indexed: 12/13/2022] Open
Abstract
Osteosarcomas, the most common malignant bone tumors, show a potent capacity for local invasion and pulmonary metastasis. Resveratrol (RESV), a phytochemical, exhibits multiple tumor-suppressing activities and has been tested in clinical trials. However, the antitumor activities of RESV in osteosarcomas are not yet completely defined. In osteosarcoma cells, we found that RESV inhibited the migration/invasion in vitro and lung metastasis in vivo by suppressing matrix metalloproteinase (MMP)-2. We identified that RESV exhibited a transcriptional inhibitory effect on MMP-2 through reducing CREB-DNA-binding activity. Moreover, a microRNA (miR) analysis showed that miR-328 was predominantly upregulated after RESV treatment. Inhibition of miR-328 significantly relieved MMP-2 and motility suppression imposed by RESV treatment. Furthermore, ectopic miR-328 expression in highly invasive cells decreased MMP-2 expression and invasive abilities. Mechanistic investigations found that JNK and p38 MAPK signaling pathways were involved in RESV-regulated CREB-DNA-binding activity, miR328 expression, and cell motility. Clinical samples indicated inverse expression between MMP-2 and miR-328 in normal bone and osteosarcoma tissues. The inverse correlation of MMP-2 and miR-328 was also observed in tumor specimens, and MMP-2 expression was linked to tumor metastasis. Taken together, our results provide new insights into the role of RESV-induced molecular and epigenetic regulation in suppressing tumor metastasis.
Collapse
Affiliation(s)
- Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Wei-Jiunn Lee
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Peng Tan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Michael Hsiao
- The Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Feng-Koo Hsieh
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians University, Munich, Germany
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
211
|
Xiang Q, Zhang J, Li CY, Wang Y, Zeng MJ, Cai ZX, Tian RB, Jia W, Li XH. Insulin resistance-induced hyperglycemia decreased the activation of Akt/CREB in hippocampus neurons: Molecular evidence for mechanism of diabetes-induced cognitive dysfunction. Neuropeptides 2015; 54:9-15. [PMID: 26344332 DOI: 10.1016/j.npep.2015.08.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/27/2015] [Accepted: 08/27/2015] [Indexed: 02/07/2023]
Abstract
Several previous studies have indicated that diabetic have higher risk of suffering from Alzheimer's disease, which severely induced cognitive dysfunction. However, the underlying molecular mechanism and more details on the cognitive deficits induced by hyperglycemia have not been elucidated. Here in our present study, on the basis of Goto-Kakizaki (GK) rats and streptozotocin (STZ)-induced diabetic model, we detected the variation of dendritic spine density in hippocampus as well as the differential expression of some important signal transduction molecules that were of relevance in learning and memory function. We found that the magnitude of escape latency time was significantly increased in such diabetic animals; the phosphorylated Akt/CREB; SYP and BDNF as well as other downstream molecules in hippocampus neurons were also downregulated in both diabetic groups compared to the normal groups. Thus, all of these data indicate the obstacle of neuronal pathology and the Akt/CREB signaling pathway caused by hyperglycemia that may suppress cognitive behavior, which may provide a novel way for the prevention of diabetic encephalopathy and the cognitive deficits of Alzheimer's disease.
Collapse
Affiliation(s)
- Qiong Xiang
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| | - Jie Zhang
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| | - Chun-Yan Li
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| | - Yan Wang
- Pharmacy of Department, First People's Hospital of Foshan, Guangdong, China
| | - Mao-Jun Zeng
- College of Medicine, Jishou University, Hunan, China
| | - Zhi-Xin Cai
- College of Medicine, Jishou University, Hunan, China
| | - Rong-Bo Tian
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| | - Wei Jia
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| | - Xian-Hui Li
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China.
| |
Collapse
|
212
|
Pesiri V, Totta P, Segatto M, Bianchi F, Pallottini V, Marino M, Acconcia F. Estrogen receptor α L429 and A430 regulate 17β-estradiol-induced cell proliferation via CREB1. Cell Signal 2015; 27:2380-8. [DOI: 10.1016/j.cellsig.2015.08.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 08/25/2015] [Accepted: 08/28/2015] [Indexed: 10/23/2022]
|
213
|
Zhang X, Wang Y, Wang J, Sun F. Protein-protein interactions among signaling pathways may become new therapeutic targets in liver cancer (Review). Oncol Rep 2015; 35:625-38. [PMID: 26717966 DOI: 10.3892/or.2015.4464] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/06/2015] [Indexed: 11/05/2022] Open
Abstract
Numerous signaling pathways have been shown to be dysregulated in liver cancer. In addition, some protein-protein interactions are prerequisite for the uncontrolled activation or inhibition of these signaling pathways. For instance, in the PI3K/AKT signaling pathway, protein AKT binds with a number of proteins such as mTOR, FOXO1 and MDM2 to play an oncogenic role in liver cancer. The aim of the present review was to focus on a series of important protein-protein interactions that can serve as potential therapeutic targets in liver cancer among certain important pro-carcinogenic signaling pathways. The strategies of how to investigate and analyze the protein-protein interactions are also included in this review. A survey of these protein interactions may provide alternative therapeutic targets in liver cancer.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, P.R. China
| | - Yulan Wang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, P.R. China
| | - Jiayi Wang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, P.R. China
| | - Fenyong Sun
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, P.R. China
| |
Collapse
|
214
|
He X, Thacker S, Romigh T, Yu Q, Frazier TW, Eng C. Cytoplasm-predominant Pten associates with increased region-specific brain tyrosine hydroxylase and dopamine D2 receptors in mouse model with autistic traits. Mol Autism 2015; 6:63. [PMID: 26579216 PMCID: PMC4647625 DOI: 10.1186/s13229-015-0056-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 11/09/2015] [Indexed: 11/13/2022] Open
Abstract
Background Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders characterized by impairment in social communication/interaction and inflexible/repetitive behavior. Several lines of evidence support genetic factors as a predominant cause of ASD. Among those autism susceptibility genes that have been identified, the PTEN tumor suppressor gene, initially identified as predisposing to Cowden heritable cancer syndrome, was found to be mutated in a subset of ASD patients with extreme macrocephaly. However, the ASD-relevant molecular mechanism mediating the effect of PTEN mutations remains elusive. Methods We developed a Pten knock-in murine model to study the effects of Pten germline mutations, specifically altering subcellular localization, in ASD. Proteins were isolated from the hemispheres of the male littermates, and Western blots were performed to determine protein expression levels of tyrosine hydroxylase (TH). Immunohistochemical stains were carried out to validate the localization of TH and dopamine D2 receptors (D2R). PC12 cells ectopically expressing either wild-type or missense mutant PTEN were then compared for the differences in TH expression. Results Mice carrying Pten mutations have high TH and D2R in the striatum and prefrontal cortex. They also have increased phosphorylation of cAMP response element-binding protein (CREB) and TH. Mechanistically, PTEN downregulates TH production in PC12 cells via inhibiting the phosphoinositide 3-kinase (PI3K)/CREB signaling pathway, while PTEN reduces TH phosphorylation via suppressing MAPK pathway. Unlike wild-type PTEN but similar to the mouse knock-in mutant Pten, three naturally occurring missense mutations of PTEN that we previously identified in ASD patients, H93R, F241S, and D252G, were not able to suppress TH when overexpressed in PC12 cells. In addition, two other PTEN missense mutations, C124S (pan phosphatase dead) and G129E (lipid phosphatase dead), failed to suppress TH when ectopically expressed in PC12 cells. Conclusions Our data reveal a non-canonical PTEN-TH pathway in the brain that may work as a core regulator of dopamine signaling, which when dysfunctional is pathogenic in ASD. Electronic supplementary material The online version of this article (doi:10.1186/s13229-015-0056-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xin He
- Genomic Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, Mailstop NE-50, Cleveland, OH 44195 USA ; Lerner Research Institute, Cleveland Clinic, Cleveland, OH USA
| | - Stetson Thacker
- Genomic Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, Mailstop NE-50, Cleveland, OH 44195 USA ; Lerner Research Institute, Cleveland Clinic, Cleveland, OH USA ; HHMI Graduate Program, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH USA
| | - Todd Romigh
- Genomic Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, Mailstop NE-50, Cleveland, OH 44195 USA ; Lerner Research Institute, Cleveland Clinic, Cleveland, OH USA
| | - Qi Yu
- Genomic Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, Mailstop NE-50, Cleveland, OH 44195 USA ; Lerner Research Institute, Cleveland Clinic, Cleveland, OH USA
| | - Thomas W Frazier
- Genomic Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, Mailstop NE-50, Cleveland, OH 44195 USA ; Lerner Research Institute, Cleveland Clinic, Cleveland, OH USA ; HHMI Graduate Program, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH USA ; Center for Autism, Pediatrics Institute, Cleveland Clinic, Cleveland, OH USA ; Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH USA
| | - Charis Eng
- Genomic Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, Mailstop NE-50, Cleveland, OH 44195 USA ; Lerner Research Institute, Cleveland Clinic, Cleveland, OH USA ; HHMI Graduate Program, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH USA ; Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH USA ; Stanley Shalom Zielony Institute of Nursing Excellence, Cleveland Clinic, Cleveland, OH USA ; Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH USA ; CASE Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH USA
| |
Collapse
|
215
|
Fröhlich H, Bahamondez G, Götschel F, Korf U. Dynamic Bayesian Network Modeling of the Interplay between EGFR and Hedgehog Signaling. PLoS One 2015; 10:e0142646. [PMID: 26571415 PMCID: PMC4646463 DOI: 10.1371/journal.pone.0142646] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/23/2015] [Indexed: 11/23/2022] Open
Abstract
Aberrant activation of sonic Hegdehog (SHH) signaling has been found to disrupt cellular differentiation in many human cancers and to increase proliferation. The SHH pathway is known to cross-talk with EGFR dependent signaling. Recent studies experimentally addressed this interplay in Daoy cells, which are presumable a model system for medulloblastoma, a highly malignant brain tumor that predominately occurs in children. Currently ongoing are several clinical trials for different solid cancers, which are designed to validate the clinical benefits of targeting the SHH in combination with other pathways. This has motivated us to investigate interactions between EGFR and SHH dependent signaling in greater depth. To our knowledge, there is no mathematical model describing the interplay between EGFR and SHH dependent signaling in medulloblastoma so far. Here we come up with a fully probabilistic approach using Dynamic Bayesian Networks (DBNs). To build our model, we made use of literature based knowledge describing SHH and EGFR signaling and integrated gene expression (Illumina) and cellular location dependent time series protein expression data (Reverse Phase Protein Arrays). We validated our model by sub-sampling training data and making Bayesian predictions on the left out test data. Our predictions focusing on key transcription factors and p70S6K, showed a high level of concordance with experimental data. Furthermore, the stability of our model was tested by a parametric bootstrap approach. Stable network features were in agreement with published data. Altogether we believe that our model improved our understanding of the interplay between two highly oncogenic signaling pathways in Daoy cells. This may open new perspectives for the future therapy of Hedghog/EGF-dependent solid tumors.
Collapse
Affiliation(s)
- Holger Fröhlich
- Algorithmic Bioinformatics, Institute for Computer Science, c/o Bonn-Aachen International Center for IT (B-IT), University of Bonn, Bonn, Germany
- * E-mail:
| | - Gloria Bahamondez
- Algorithmic Bioinformatics, Institute for Computer Science, c/o Bonn-Aachen International Center for IT (B-IT), University of Bonn, Bonn, Germany
| | - Frank Götschel
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrike Korf
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
216
|
Bak DH, Zhang E, Yi MH, Kim DK, Lim K, Kim JJ, Kim DW. High ω3-polyunsaturated fatty acids in fat-1 mice prevent streptozotocin-induced Purkinje cell degeneration through BDNF-mediated autophagy. Sci Rep 2015; 5:15465. [PMID: 26503303 PMCID: PMC4621527 DOI: 10.1038/srep15465] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 09/17/2015] [Indexed: 12/14/2022] Open
Abstract
Loss of Purkinje cells has been implicated in the development of diabetic neuropathy, and this degeneration is characterized by impairment of autophagic processes. We evaluated whether fat-1 transgenic mice, a well-established animal model that endogenously synthesizes ω3 polyunsaturated fatty acids (ω3-PUFA), are protected from Purkinje cell degeneration in streptozotocin (STZ)-treated model with fat-1 mice. STZ-treated fat-1 mice did not develop hyperglycemia, motor deficits, or Purkinje cell loss. The expression of LC3 I, II, Beclin-1 and p62 were increased in the cerebellum of STZ-treated wild-type mice, and these expressions were more increased in STZ-treated fat-1 mice, but not of p62. Moreover, cerebellar Rab7, Cathepsin D, and ATP6E were increased in STZ-treated fat-1 mice. There was also increased BDNF expression in Purkinje cells without any changes in TrkB, and phosphorylation of Akt and CREB in the cerebellums of fat-1 mice. Collectively, these findings indicate that STZ-treated fat-1 mice were protected from Purkinje cell loss and exhibited increased BDNF signaling, enhancing autophagic flux activity in cerebellar Purkinje neurons. These processes may underlie Purkinje cell survival and may be potential therapeutic targets for treatment of motor deficits related to diabetic neuropathy.
Collapse
Affiliation(s)
- Dong Ho Bak
- Department of Anatomy, College of Medicine, Konyang University of Korea, Daejeon, South Korea
| | - Enji Zhang
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Anesthesiology, Yanbian University Hospital, Yanbian, 133000, China
| | - Min-Hee Yi
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Do-Kyung Kim
- Department of Pharmacology, College of Medicine, Konyang University of Korea, Daejeon, South Korea
| | - Kyu Lim
- Department of Biochemistry, Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Jwa-Jin Kim
- Department of Anatomy, College of Medicine, Konyang University of Korea, Daejeon, South Korea
| | - Dong Woon Kim
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, South Korea
| |
Collapse
|
217
|
Ehrlich DE, Josselyn SA. Plasticity-related genes in brain development and amygdala-dependent learning. GENES BRAIN AND BEHAVIOR 2015; 15:125-43. [PMID: 26419764 DOI: 10.1111/gbb.12255] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/12/2015] [Accepted: 09/14/2015] [Indexed: 12/31/2022]
Abstract
Learning about motivationally important stimuli involves plasticity in the amygdala, a temporal lobe structure. Amygdala-dependent learning involves a growing number of plasticity-related signaling pathways also implicated in brain development, suggesting that learning-related signaling in juveniles may simultaneously influence development. Here, we review the pleiotropic functions in nervous system development and amygdala-dependent learning of a signaling pathway that includes brain-derived neurotrophic factor (BDNF), extracellular signaling-related kinases (ERKs) and cyclic AMP-response element binding protein (CREB). Using these canonical, plasticity-related genes as an example, we discuss the intersection of learning-related and developmental plasticity in the immature amygdala, when aversive and appetitive learning may influence the developmental trajectory of amygdala function. We propose that learning-dependent activation of BDNF, ERK and CREB signaling in the immature amygdala exaggerates and accelerates neural development, promoting amygdala excitability and environmental sensitivity later in life.
Collapse
Affiliation(s)
- D E Ehrlich
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, NY, USA.,Department of Otolaryngology, NYU Langone School of Medicine, New York, NY, USA
| | - S A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, ON, Canada.,Department of Psychology, University of Toronto, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
218
|
Chen J, Harris RC. Interaction of the EGF Receptor and the Hippo Pathway in the Diabetic Kidney. J Am Soc Nephrol 2015; 27:1689-700. [PMID: 26453611 DOI: 10.1681/asn.2015040415] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 08/27/2015] [Indexed: 12/30/2022] Open
Abstract
Activation of the EGF receptor (EGFR) or the Hippo signaling pathway can control cell proliferation, apoptosis, and differentiation, and the dysregulation of these pathways can contribute to tumorigenesis. Previous studies showed that activation of EGFR signaling in renal epithelial cells can exacerbate diabetic kidney injury. Moreover, EGFR has been implicated in regulating the Hippo signaling pathway in Drosophila; thus, we examined this potential interaction in mammalian diabetic kidney disease. Yes-associated protein (YAP) is a transcriptional regulator regulated by the Hippo signaling pathway. We found YAP protein expression and phosphorylation were upregulated in diabetic mouse renal proximal tubule epithelial cells, which were inhibited in diabetic proximal tubule EGFR-knockout mice (EGFR(ptKO)) or administration of an EGFR tyrosine kinase inhibitor erlotinib. Furthermore, activation of an EGFR-PI3K-Akt-CREB signaling pathway mediated YAP gene expression and YAP nuclear translocation and interaction with the TEA domain (TEAD) transcription factor complex, which led to upregulated expression of two TEAD-dependent genes, the connective tissue growth factor and amphiregulin genes. In a renal proximal tubule cell line, either pharmacologic or genetic inhibition of EGFR, Akt, or CREB blunted YAP expression in response to high-glucose treatment. Additionally, knocking down YAP expression by specific siRNA inhibited cell proliferation in response to high glucose or exogenous EGF. Therefore, these results link the Hippo pathway to EGFR-mediated renal epithelial injury in diabetes.
Collapse
Affiliation(s)
- Jianchun Chen
- Department of Veterans Affairs, Nashville, Tennessee; and Department of Medicine and
| | - Raymond C Harris
- Department of Veterans Affairs, Nashville, Tennessee; and Department of Medicine and Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
219
|
Ortega-Martínez S. A new perspective on the role of the CREB family of transcription factors in memory consolidation via adult hippocampal neurogenesis. Front Mol Neurosci 2015; 8:46. [PMID: 26379491 PMCID: PMC4549561 DOI: 10.3389/fnmol.2015.00046] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 08/11/2015] [Indexed: 12/21/2022] Open
Abstract
Adult neurogenesis is the process by which new neurons are generated in the brains of adults. Since its discovery 50 years ago, adult neurogenesis has been widely studied in the mammalian brain and has provided a new perspective on the pathophysiology of many psychiatric and neurodegenerative disorders, some of which affect memory. In this regard, adult hippocampal neurogenesis (AHN), which occurs in the subgranular zone (SGZ) of the dentate gyrus (DG), has been suggested to play a role in the formation and consolidation of new memories. This process involves many transcription factors, of which cyclic AMP (cAMP)-responsive element-binding protein (CREB) is a well-documented one. In the developing brain, CREB regulates crucial cell stages (e.g., proliferation, differentiation, and survival), and in the adult brain, it participates in neuronal plasticity, learning, and memory. In addition, new evidence supports the hypothesis that CREB may also participate in learning and memory through its involvement in AHN. This review examines the CREB family of transcription factors, including the different members and known signaling pathways. It highlights the role of CREB as a modulator of AHN, which could underlie its function in memory consolidation mechanisms.
Collapse
Affiliation(s)
- Sylvia Ortega-Martínez
- Turku Centre for Biotechnology, Åbo Akademi University and University of Turku Turku, Finland
| |
Collapse
|
220
|
Lee CJ, Jang JH, Lee JY, Lee MH, Li Y, Ryu HW, Choi KI, Dong Z, Lee HS, Oh SR, Surh YJ, Cho YY. Aschantin targeting on the kinase domain of mammalian target of rapamycin suppresses epidermal growth factor-induced neoplastic cell transformation. Carcinogenesis 2015; 36:1223-34. [PMID: 26243309 DOI: 10.1093/carcin/bgv113] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/27/2015] [Indexed: 12/22/2022] Open
Abstract
Mammalian target of rapamycin (mTOR), a serine/threonine protein kinase, forms two different complexes, complex 1 and 2, and plays a key role in the regulation of Akt signaling-mediated cell proliferation and transformation. This study reveals aschantin, a natural compound abundantly found in Magnolia flos, as a novel mTOR kinase inhibitor. Aschantin directly targeted the active pocket of mTOR kinase domain by competing with adenosine triphosphate (ATP), but not PI3K and PDK1. Aschantin inhibited epidermal growth factor (EGF)-induced full activation of Akt by phosphorylation at Ser473/Thr308, resulting in inhibition of the mTORC2/Akt and Akt/mTORC1/p70S6K signaling pathways and activation of GSK3β by abrogation of Akt-mediated GSK3β phosphorylation at Ser9. The activated GSK3β inhibited cell proliferation by c-Jun phosphorylation at Ser243, which facilitated destabilization and degradation of c-Jun through the ubiquitination-mediated proteasomal degradation pathway. Notably, aschantin treatment decreased c-Jun stability through inhibition of the mTORC2-Akt signaling pathway, which suppressed EGF-induced anchorage-independent cell transformation in non-malignant JB6 Cl41 and HaCaT cells and colony growth of LNCaP and MIAPaCa-2 cancer cells in soft agar. Altogether, the results show that aschantin targets mTOR kinase and destabilizes c-Jun, which implicate aschantin as a potential chemopreventive or therapeutic agent.
Collapse
Affiliation(s)
- Cheol-Jung Lee
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 420-743, Republic of Korea
| | - Jeong-Hoon Jang
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 420-743, Republic of Korea, College of Pharmacy, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Ji-Young Lee
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 420-743, Republic of Korea
| | - Mee-Hyun Lee
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 420-743, Republic of Korea
| | - Yan Li
- The Hormel Institute, University of Minnesota, 801, 16th AVE, NE, Austin, MN 55912, USA and
| | - Hyung Won Ryu
- Natural Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gun, ChungBuk 363-883, Republic of Korea
| | - Kyung-Il Choi
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 420-743, Republic of Korea
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, 801, 16th AVE, NE, Austin, MN 55912, USA and
| | - Hye Suk Lee
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 420-743, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gun, ChungBuk 363-883, Republic of Korea
| | - Young-Joon Surh
- College of Pharmacy, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul 151-742, Republic of Korea,
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 420-743, Republic of Korea,
| |
Collapse
|
221
|
Bavithra S, Sugantha Priya E, Selvakumar K, Krishnamoorthy G, Arunakaran J. Effect of Melatonin on Glutamate: BDNF Signaling in the Cerebral Cortex of Polychlorinated Biphenyls (PCBs)-Exposed Adult Male Rats. Neurochem Res 2015. [PMID: 26224288 DOI: 10.1007/s11064-015-1677-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Various epidemiological survey suggests that the central nervous system is the target for many environmental contaminants. One among them is Aroclor 1254, a mixture of polychlorinated biphenyls (PCBs) which explore a spectrum of biochemical and neurotoxic responses in humans and laboratory animals. Learning and motor coordination deficits are the profound effects of PCBs which may be related to cerebral dysfunction. The aim of the study is to elicit the protective effect of melatonin (Mel), a potent, blood brain permeable antioxidant against the effect of Aroclor 1254 on the signaling of glutamate-principal excitatory neurotransmitter and brain derived neurotrophic factor (BDNF) in the cerebral cortex of adult rats which plays a key role in brain functions. Adult male Wistar rats were grouped into four and treated intraperitonealy (i.p) Group I with corn oil (Control), Group II with PCBs (2 mg/kg/bwt), Group III with PCBs + Mel (2 mg/kg/bwt + 5 mg/kg/bwt) and Group IV with Mel (5 mg/kg/bwt). The protein expression of glutamate signaling molecules and mRNA expressions of GLAST, BDNF signaling molecules were analyzed. The results suggest that simultaneous melatonin treatment significantly attenuated the NMDA receptor mediated glutamate excitotoxicity and protects the inhibition of BDNF signaling caused by PCBs exposure in cerebral cortex of adult male rats. Schematic pathway illustrating the proposed mechanism by which melatonin protects against A1254 mediated glutamate induced neurodegeneration in the cerebral cortex of adult male rats. PCBs induced neurodegeneration is caused by the overactivation of NMDAR, followed by the activation of voltage dependent calcium channels leading to the increase in intracellular Ca(2+) that stimulates calpain. Calpain inturn inhibits the PKA α and neurtrophin BDNF, its receptor and downstream signaling MAPK pathway leading to neurodegeneration. Melatonin had scavenged the ROS produced by PCBS and decreased the NMDAR expression which inturn protected the cells from neurodegeneration.
Collapse
Affiliation(s)
- S Bavithra
- Department of Endocrinology, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai, 600 113, India
| | | | | | | | | |
Collapse
|
222
|
Zhang S, Patel A, Moorthy B, Shivanna B. Adrenomedullin deficiency potentiates hyperoxic injury in fetal human pulmonary microvascular endothelial cells. Biochem Biophys Res Commun 2015. [PMID: 26196743 DOI: 10.1016/j.bbrc.2015.07.067] [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] [Indexed: 02/06/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease of premature infants that is characterized by alveolar simplification and decreased lung angiogenesis. Hyperoxia-induced oxidative stress and inflammation contributes to the development of BPD in premature infants. Adrenomedullin (AM) is an endogenous peptide with potent angiogenic, anti-oxidant, and anti-inflammatory properties. Whether AM regulates hyperoxic injury in fetal primary human lung cells is unknown. Therefore, we tested the hypothesis that AM-deficient fetal primary human pulmonary microvascular endothelial cells (HPMEC) will have increased oxidative stress, inflammation, and cytotoxicity compared to AM-sufficient HPMEC upon exposure to hyperoxia. Adrenomedullin gene (Adm) was knocked down in HPMEC by siRNA-mediated transfection and the resultant AM-sufficient and -deficient cells were evaluated for hyperoxia-induced oxidative stress, inflammation, cytotoxicity, and Akt activation. AM-deficient HPMEC had significantly increased hyperoxia-induced reactive oxygen species (ROS) generation and cytotoxicity compared to AM-sufficient HPMEC. Additionally, AM-deficient cell culture supernatants had increased macrophage inflammatory protein 1α and 1β, indicating a heightened inflammatory state. Interestingly, AM deficiency was associated with an abrogated Akt activation upon exposure to hyperoxia. These findings support the hypothesis that AM deficiency potentiates hyperoxic injury in primary human fetal HPMEC via mechanisms entailing Akt activation.
Collapse
Affiliation(s)
- Shaojie Zhang
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Ananddeep Patel
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Bhagavatula Moorthy
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Binoy Shivanna
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
223
|
Chang H, Jung WY, Kang Y, Lee H, Kim A, Kim BH. Expression of ROR1, pAkt, and pCREB in gastric adenocarcinoma. Ann Diagn Pathol 2015; 19:330-4. [PMID: 26245996 DOI: 10.1016/j.anndiagpath.2015.06.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/27/2015] [Accepted: 06/24/2015] [Indexed: 10/23/2022]
Abstract
The receptor tyrosine kinase-like orphan receptor 1 (ROR1) is a transmembrane protein of receptor tyrosine kinase family. High expression of ROR1 is reported in many types of malignancies and is thought to be involved in tumor growth, apoptosis, and epithelial-mesenchymal transition. In this study, we examined the expression of ROR1, pAkt, and pCREB in gastric adenocarcinoma and analyzed with clinicopathologic factors and tumor proliferation. Tissue microarray blocks containing 424 gastric adenocarcinomas were used for immunohistochemical staining. Ki-67 labeling index was used for tumor proliferation activity. High expression of ROR1 (63%), pAkt (36%), and pCREB (20%) was observed in gastric adenocarcinomas, and expression of these proteins was well intercorrelated. ROR1 and pCREB expression was associated with Ki-67 labeling index (P < .001). Expression of pAkt and pCREB group showed longer survival in univariate analysis (P = .007 and P < .001, respectively). This is the first study that analyzed ROR1 expression in gastric adenocarcinoma tissue samples. We revealed that gastric adenocarcinomas highly express ROR1 and related proteins and its prognostic significance. ROR1 in gastric adenocarcinoma could be possible candidate of therapeutic target, and more comprehensive study is required.
Collapse
Affiliation(s)
- Hyeyoon Chang
- Department of Pathology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Woon Yong Jung
- Department of Pathology, Catholic Kwandong University International St Mary's Hospital, Incheon, Republic of Korea
| | - Youngran Kang
- Department of Pathology, Green Cross Laboratories, Yongin, Kyeonggi-Do, Republic of Korea
| | - Hyunjoo Lee
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University College of Medicine, Seoul, Republic of Korea
| | - Aeree Kim
- Department of Pathology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Baek-hui Kim
- Department of Pathology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
224
|
Habib SL, Mohan S, Liang S, Li B, Yadav M. Novel mechanism of transcriptional regulation of cell matrix protein through CREB. Cell Cycle 2015; 14:2598-608. [PMID: 26115221 DOI: 10.1080/15384101.2015.1064204] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The transcription mechanism(s) of renal cell matrix accumulation in diabetes does not explored. Phosphorylation of the transcription factor cAMP-responsive element binding protein (CREB) significantly increased in cells treated with high glucose (HG) compared to cell grown in normal glucose (NG). Cells pretreated with rapamycin before exposure to HG showed significant decrease phosphorylation of CREB, increase in AMPK activity and decrease protein/mRNA and promoter activity of fibronectin. In addition, cells transfected with siRNA against CREB showed significant increase in AMPK activity, decrease in protein/mRNA and promoter activity of fibronectin. Cells treated with HG showed nuclear localization of p-CREB while pretreated cells with rapamycin reversed HG effect. Moreover, gel shift analysis shows increase binding of CREB to fibronectin promoter in cells treated with HG while cells pretreated with rapamycin reversed the effect of HG. Furthermore, db/db mice treated with rapamycin showed significant increase in AMPK activity, decrease in expression of p-CREB and protein/mRNA of fibronectin. Strong staining of fibronectin and p-CREB was detected in kidney cortex of db/db mice while treated mice with rapamycin reversed hyperglycemia effect. In summary, our data provide a novel mechanism of transcriptional regulation of fibronectin through CREB that may be used as therapeutic approach to prevent diabetes complications.
Collapse
Affiliation(s)
- Samy L Habib
- a South Texas Veterans Health Care System ; San Antonio , TX USA
| | | | | | | | | |
Collapse
|
225
|
Signaling Circuits and Regulation of Immune Suppression by Ovarian Tumor-Associated Macrophages. Vaccines (Basel) 2015; 3:448-66. [PMID: 26343197 PMCID: PMC4494355 DOI: 10.3390/vaccines3020448] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 05/24/2015] [Accepted: 05/26/2015] [Indexed: 12/25/2022] Open
Abstract
The barriers presented by immune suppression in the ovarian tumor microenvironment present one of the biggest challenges to development of successful tumor vaccine strategies for prevention of disease recurrence and progression following primary surgery and chemotherapy. New insights gained over the last decade have revealed multiple mechanisms of immune regulation, with ovarian tumor-associated macrophages/DC likely to fulfill a central role in creating a highly immunosuppressive milieu that supports disease progression and blocks anti-tumor immunity. This review provides an appraisal of some of the key signaling pathways that may contribute to immune suppression in ovarian cancer, with a particular focus on the potential involvement of the c-KIT/PI3K/AKT, wnt/β-catenin, IL-6/STAT3 and AhR signaling pathways in regulation of indoleamine 2,3-dioxygenase expression in tumor-associated macrophages. Knowledge of intercellular and intracellular circuits that shape immune suppression may afford insights for development of adjuvant treatments that alleviate immunosuppression in the tumor microenvironment and enhance the clinical efficacy of ovarian tumor vaccines.
Collapse
|
226
|
Fröhlich D, Kuo WP, Frühbeis C, Sun JJ, Zehendner CM, Luhmann HJ, Pinto S, Toedling J, Trotter J, Krämer-Albers EM. Multifaceted effects of oligodendroglial exosomes on neurons: impact on neuronal firing rate, signal transduction and gene regulation. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0510. [PMID: 25135971 DOI: 10.1098/rstb.2013.0510] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Exosomes are small membranous vesicles of endocytic origin that are released by almost every cell type. They exert versatile functions in intercellular communication important for many physiological and pathological processes. Recently, exosomes attracted interest with regard to their role in cell-cell communication in the nervous system. We have shown that exosomes released from oligodendrocytes upon stimulation with the neurotransmitter glutamate are internalized by neurons and enhance the neuronal stress tolerance. Here, we demonstrate that oligodendroglial exosomes also promote neuronal survival during oxygen-glucose deprivation, a model of cerebral ischaemia. We show the transfer from oligodendrocytes to neurons of superoxide dismutase and catalase, enzymes which are known to help cells to resist oxidative stress. Additionally, we identify various effects of oligodendroglial exosomes on neuronal physiology. Electrophysiological analysis using in vitro multi-electrode arrays revealed an increased firing rate of neurons exposed to oligodendroglial exosomes. Moreover, gene expression analysis and phosphorylation arrays uncovered differentially expressed genes and altered signal transduction pathways in neurons after exosome treatment. Our study thus provides new insight into the broad spectrum of action of oligodendroglial exosomes and their effects on neuronal physiology. The exchange of extracellular vesicles between neural cells may exhibit remarkable potential to impact brain performance.
Collapse
Affiliation(s)
- Dominik Fröhlich
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany
| | - Wen Ping Kuo
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany Focus Programme Translational Neuroscience, University of Mainz, 55128 Mainz, Germany
| | - Carsten Frühbeis
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany
| | - Jyh-Jang Sun
- Institute of Physiology, University Medical Center, 55128 Mainz, Germany Neuro-Electronics Research Flanders, 3001 Leuven, Belgium
| | - Christoph M Zehendner
- Institute of Physiology, University Medical Center, 55128 Mainz, Germany ZIM III, Department of Cardiology, Goethe University Frankfurt, 60389 Frankfurt, Germany
| | - Heiko J Luhmann
- Focus Programme Translational Neuroscience, University of Mainz, 55128 Mainz, Germany Institute of Physiology, University Medical Center, 55128 Mainz, Germany
| | - Sheena Pinto
- Division of Developmental Immunology, DKFZ Heidelberg, 69120 Heidelberg, Germany
| | - Joern Toedling
- Institute of Molecular Biology gGmbH, 55128 Mainz, Germany
| | - Jacqueline Trotter
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany Focus Programme Translational Neuroscience, University of Mainz, 55128 Mainz, Germany
| | - Eva-Maria Krämer-Albers
- Molecular Cell Biology, University of Mainz, 55128 Mainz, Germany Focus Programme Translational Neuroscience, University of Mainz, 55128 Mainz, Germany
| |
Collapse
|
227
|
Cherry AD, Piantadosi CA. Regulation of mitochondrial biogenesis and its intersection with inflammatory responses. Antioxid Redox Signal 2015; 22:965-76. [PMID: 25556935 PMCID: PMC4390030 DOI: 10.1089/ars.2014.6200] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Mitochondria play a vital role in cellular homeostasis and are susceptible to damage from inflammatory mediators released by the host defense. Cellular recovery depends, in part, on mitochondrial quality control programs, including mitochondrial biogenesis. RECENT ADVANCES Early-phase inflammatory mediator proteins interact with PRRs to activate NF-κB-, MAPK-, and PKB/Akt-dependent pathways, resulting in increased expression or activity of coactivators and transcription factors (e.g., PGC-1α, NRF-1, NRF-2, and Nfe2l2) that regulate mitochondrial biogenesis. Inflammatory upregulation of NOS2-induced NO causes mitochondrial dysfunction, but NO is also a signaling molecule upregulating mitochondrial biogenesis via PGC-1α, participating in Nfe2l2-mediated antioxidant gene expression and modulating inflammation. NO and reactive oxygen species generated by the host inflammatory response induce the redox-sensitive HO-1/CO system, causing simultaneous induction of mitochondrial biogenesis and antioxidant gene expression. CRITICAL ISSUES Recent evidence suggests that mitochondrial biogenesis and mitophagy are coupled through redox pathways; for instance, parkin, which regulates mitophagy in chronic inflammation, may also modulate mitochondrial biogenesis and is upregulated through NF-κB. Further research on parkin in acute inflammation is ongoing. This highlights certain common features of the host response to acute and chronic inflammation, but caution is warranted in extrapolating findings across inflammatory conditions. FUTURE DIRECTIONS Inflammatory mitochondrial dysfunction and oxidative stress initiate further inflammatory responses through DAMP/PRR interactions and by inflammasome activation, stimulating mitophagy. A deeper understanding of mitochondrial quality control programs' impact on intracellular inflammatory signaling will improve our approach to the restoration of mitochondrial homeostasis in the resolution of acute inflammation.
Collapse
Affiliation(s)
- Anne D Cherry
- 1 Department of Anesthesiology, Duke University Medical Center , Durham, North Carolina
| | | |
Collapse
|
228
|
Holenya P, Can S, Rubbiani R, Alborzinia H, Jünger A, Cheng X, Ott I, Wölfl S. Detailed analysis of pro-apoptotic signaling and metabolic adaptation triggered by a N-heterocyclic carbene-gold(I) complex. Metallomics 2015; 6:1591-601. [PMID: 24777153 DOI: 10.1039/c4mt00075g] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Due to their broad spectrum of biological activity and antiproliferative effect on different human cancer cell lines, gold compounds have been in the focus of drug research for many years. Gold(I)-N-heterocyclic carbene complexes are of particular interest, because of their stability, ease of derivatization and clear cytotoxicity in cancer cells. To obtain a more detailed view of the molecular mechanisms underlying their cellular activity, we used a novel gold(I)-N-heterocyclic carbene complex, [triphenylphosphane-(1,3-diethyl-5-methoxy-benzylimidazol-2-ylidene)]gold(I) iodide and investigated changes in cellular signaling pathways using quantitative signal transduction protein microarray analysis. We also analyzed changes in cell metabolism in a time-dependent manner by on-line metabolic measurements and used isolated mitochondria to elucidate the direct effects on this cell organelle. We found strong cytotoxic effects in cancer cells, accompanied by an immediate and irreversible loss of mitochondrial respiration as well as by a crucial imbalance of the intracellular redox state, resulting in apoptotic cell death. ELISA microarray analysis of signal transduction pathways revealed a time-dependent up-regulation of pro-apoptotic signaling proteins, e.g. p38 and JNK, whereas pro-survival signals that are directly linked to the thioredoxin system were down-regulated, which pinpoints to thioredoxin reductase as a central target of the compound. Further results suggest that DNA is an indirect target of the compound. Based on our findings, we outline a signaling model for the molecular mechanism underlying the antiproliferative activity of the gold(I)-N-heterocyclic carbene complex investigated, which provides a good general model for the known pattern of cell death induced by this class of substances.
Collapse
Affiliation(s)
- Pavlo Holenya
- Department of Biology, Institut für Pharmazie und molekulare Biotechnologie, Ruperto-Carola University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
229
|
Breivik L, Jensen A, Guvåg S, Aarnes EK, Aspevik A, Helgeland E, Hovland S, Brattelid T, Jonassen AK. B-type natriuretic peptide expression and cardioprotection is regulated by Akt dependent signaling at early reperfusion. Peptides 2015; 66:43-50. [PMID: 25698234 DOI: 10.1016/j.peptides.2015.01.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 01/09/2015] [Accepted: 01/13/2015] [Indexed: 01/16/2023]
Abstract
Exogenously administered B-type natriuretic peptide (BNP) has been shown to offer cardioprotection through activation of particulate guanylyl cyclase (pGC), protein kinase G (PKG) and KATP channel opening. The current study explores if cardioprotection afforded by short intermittent BNP administration involves PI3K/Akt/p70s6k dependent signaling, and whether this signaling pathway may participate in regulation of BNP mRNA expression at early reperfusion. Isolated Langendorff perfused rat hearts were subjected to 30min of regional ischemia and 120min of reperfusion (IR). Applying intermittent 3×30s infusion of BNP peptide in a postconditioning like manner (BNPPost) reduced infarct size by >50% compared to controls (BNPPost 17±2% vs. control 42±4%, p<0.001). Co-treatment with inhibitors of the PI3K/Akt/p70s6k pathway (wortmannin, SH-6 and rapamycin) completely abolished the infarct-limiting effect of BNP postconditioning (BNPPost+Wi 36±5%, BNPPost+SH-6 41±4%, BNPPost+Rap 37±6% vs. BNPPost 17±2%, p<0.001). Inhibition of natriuretic peptide receptors (NPR) by isatin also abrogated BNPPost cardioprotection (BNPPost+isatin 46±2% vs. BNPPost 17±2%, p<0.001). BNPPost also significantly phosphorylated Akt and p70s6k at early reperfusion, and Akt phosphorylation was inhibited by SH-6 and isatin. Myocardial BNP mRNA levels in the area at risk (AA) were significantly elevated at early reperfusion as compared to the non-ischemic area (ANA) (Ctr(AA) 2.7±0.5 vs. Ctr(ANA) 1.2±0.2, p<0.05) and the ischemic control tissue (Ctr(AA) 2.7±0.5 vs. ischemia 1.0±0.1, p<0.05). Additional experiments also revealed a significant higher BNP mRNA level in ischemic postconditioned (IPost) hearts as compared to ischemic controls (IPost 6.7±1.3 vs. ischemia 1.0±0.2, p<0.05), but showed no difference from controls run in parallel (Ctr 5.4±0.8). Akt inhibition by SH-6 completely abrogated this elevation (IPost 6.7±1.3 vs. IPost+SH-6 1.8±0.7, p<0.05) (Ctr 5.4±0.8 vs. SH-6 1.5±0.9, p<0.05). In conclusion, Akt dependent signaling is involved in mediating the cardioprotection afforded by intermittent BNP infusion at early reperfusion, and may also participate in regulation of reperfusion induced BNP expression.
Collapse
Affiliation(s)
- L Breivik
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway.
| | - A Jensen
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - S Guvåg
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - E K Aarnes
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - A Aspevik
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - E Helgeland
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - S Hovland
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - T Brattelid
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| | - A K Jonassen
- Department of Biomedicine, Faculty of Medicine and Dentistry, University of Bergen, Norway
| |
Collapse
|
230
|
Erythropoietin activates the phosporylated cAMP [adenosine 3′5′ cyclic monophosphate] response element-binding protein pathway and attenuates delayed paraplegia after ischemia-reperfusion injury. J Thorac Cardiovasc Surg 2015; 149:920-4. [DOI: 10.1016/j.jtcvs.2014.11.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 10/20/2014] [Accepted: 11/04/2014] [Indexed: 11/30/2022]
|
231
|
Tsigelny IF, Kouznetsova VL, Jiang P, Pingle SC, Kesari S. Hierarchical control of coherent gene clusters defines the molecular mechanisms of glioblastoma. MOLECULAR BIOSYSTEMS 2015; 11:1012-28. [PMID: 25648506 DOI: 10.1039/c5mb00007f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Glioblastoma is a highly-aggressive and rapidly-lethal tumor characterized by resistance to therapy. Although data on multiple genes, proteins, and pathways are available, the key challenge is deciphering this information and identifying central molecular targets. Therapeutically targeting individual molecules is often unsuccessful due to the presence of compensatory and redundant pathways, and crosstalk. A systems biology approach that involves a hierarchical gene group networks analysis can delineate the coherent functions of different disease mediators. Here, we report an integrative networks-based analysis to identify a system of coherent gene modules in primary and secondary glioblastoma. Our study revealed a hierarchical transcriptional control of genes in these modules. We elucidated those modules responsible for conversion of the glioma-associated microglia/macrophages into glioma-supportive, immunosuppressive cells. Further, we identified clusters comprising mediators of angiogenesis, proliferation, and cell death for both primary and secondary glioblastomas. Data obtained for these clusters point to a possible role of transcription regulators that function as the gene modules mediators in glioblastoma pathogenesis. We elucidated a set of possible transcription regulators that can be targeted to affect the selected gene clusters at specific levels for glioblastoma. Our innovative approach to construct informative disease models may hold the key to successful management of complex diseases including glioblastoma and other cancers.
Collapse
Affiliation(s)
- Igor F Tsigelny
- Department of Neurosciences, University of California San Diego, 9500 Gilman Dr., MSC 0752, La Jolla, CA 92093-0752, USA.
| | | | | | | | | |
Collapse
|
232
|
Caldarelli I, Speranza MC, Bencivenga D, Tramontano A, Borgia A, Pirozzi AVA, Perrotta S, Oliva A, Della Ragione F, Borriello A. Resveratrol mimics insulin activity in the adipogenic commitment of human bone marrow mesenchymal stromal cells. Int J Biochem Cell Biol 2015; 60:60-72. [PMID: 25562512 DOI: 10.1016/j.biocel.2014.12.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 11/07/2014] [Accepted: 12/22/2014] [Indexed: 01/01/2023]
Abstract
Bone marrow mesenchymal stromal cells (BM-MSCs) are multipotent cells capable of differentiating toward osteoblatic and adipocytic phenotypes. BM-MSCs play several key roles including bone remodeling, establishment of hematopoietic niche and immune tolerance induction. Here, we investigated the effect of resveratrol (RSV), a therapeutically promising natural polyphenol, on the commitment of human BM-MSCs primary cultures. Cell differentiation was evaluated by means of morphological analysis, specific staining and expression of osteogenic and adipocytic master genes (Runx-2, PPARγ). To maintain BM-MSC multipotency, all experiments were performed on cells at very early passages. At any concentration RSV, added to standard medium, did not affect the phenotype of confluent BM-MSCs, while, when added to osteogenic or adipogenic medium, 1 μM RSV enhances the differentiation toward osteoblasts or adipocytes, respectively. Conversely, the addition of higher RSV concentration (25 μM) to both differentiation media resulted exclusively in BM-MSCs adipogenesis. Surprisingly, the analysis of RSV molecular effects demonstrated that the compound completely substitutes insulin, a key component of adipogenic medium. We also observed that RSV treatment is associated to enhanced phosphorylation of CREB, a critical effector of insulin adipogenic activity. Finally, our observations contribute to the mechanistic elucidation of the well-known RSV positive effect on insulin sensitivity and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Ilaria Caldarelli
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Maria Carmela Speranza
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Debora Bencivenga
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Annunziata Tramontano
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Alessia Borgia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | | | - Silverio Perrotta
- Dipartimento della Donna, del Bambino e di Chirurgia Generale e Specialistica, Second University of Naples, Naples, Italy
| | - Adriana Oliva
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Fulvio Della Ragione
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy.
| | - Adriana Borriello
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy.
| |
Collapse
|
233
|
Park SJ, Jin ML, An HK, Kim KS, Ko MJ, Kim CM, Choi YW, Lee YC. Emodin induces neurite outgrowth through PI3K/Akt/GSK-3β-mediated signaling pathways in Neuro2a cells. Neurosci Lett 2015; 588:101-7. [PMID: 25562207 DOI: 10.1016/j.neulet.2015.01.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 12/31/2014] [Accepted: 01/02/2015] [Indexed: 01/23/2023]
Abstract
In this study, a neurite outgrowth-inducing substance was isolated from the ethylacetate extract of the Polygonum multiflorum roots and identified as emodin by gas-liquid chromatography-mass spectrometry and (1)H NMR and (13)C NMR. Emodin displayed remarkable neurite outgrowth-inducing activity in Neuro2a cells, as demonstrated by morphological changes and immunocytochemistry for class III β-tubulin. Emodin exhibited a stronger neutrophic activity than retinoic acid (RA) known as inducer of neurite outgrowth in Neuro2a cells. Emodin treatment resulted in marked increases in phosphorylation of Akt a direct downstream signaling molecule of phosphatidylinositol 3-kinase (PI3K), but upstream of glycogen synthase kinase-3β (GSK-3β) and cAMP response element-binding protein (CREB). These augmentations and neurite-bearing cells induced by emodin were remarkably reduced by the addition of PI3K inhibitor LY294002. These results demonstrate that emodin induces neuronal differentiation of Neuro2a cells via PI3K/Akt/GSK-3β pathway.
Collapse
Affiliation(s)
- Shin-Ji Park
- College of Natural Resources and Life Science, Dong-A University, Busan 604-714, South Korea
| | - Mei Ling Jin
- Department of Horticultural Bioscience, Pusan National University, Miryang 627-706, South Korea
| | - Hyun-Kyu An
- College of Natural Resources and Life Science, Dong-A University, Busan 604-714, South Korea
| | - Kyoung-Sook Kim
- College of Natural Resources and Life Science, Dong-A University, Busan 604-714, South Korea
| | - Min Jung Ko
- Department of Horticultural Bioscience, Pusan National University, Miryang 627-706, South Korea
| | - Cheol Min Kim
- Research Center for Anti-Aging Technology Development, Busan National University, Busan 609-735, South Korea
| | - Young Whan Choi
- Department of Horticultural Bioscience, Pusan National University, Miryang 627-706, South Korea.
| | - Young-Choon Lee
- College of Natural Resources and Life Science, Dong-A University, Busan 604-714, South Korea.
| |
Collapse
|
234
|
Wan Y, Wang Q, Prud’homme GJ. GABAergic system in the endocrine pancreas: a new target for diabetes treatment. Diabetes Metab Syndr Obes 2015; 8:79-87. [PMID: 25678807 PMCID: PMC4322886 DOI: 10.2147/dmso.s50642] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Excessive loss of functional pancreatic β-cell mass, mainly due to apoptosis, is a major factor in the development of hyperglycemia in both type 1 and type 2 diabetes (T1D and T2D). In T1D, β-cells are destroyed by immunological mechanisms. In T2D, while metabolic factors are known to contribute to β-cell failure and subsequent apoptosis, mounting evidence suggests that islet inflammation also plays an important role in the loss of β-cell mass. Therefore, it is of great importance for clinical intervention to develop new therapies. γ-Aminobutyric acid (GABA), a major neurotransmitter, is also produced by islet β-cells, where it functions as an important intraislet transmitter in regulating islet-cell secretion and function. Importantly, recent studies performed in rodents, including in vivo studies of xenotransplanted human islets, reveal that GABA exerts β-cell regenerative effects. Moreover, it protects β-cells against apoptosis induced by cytokines, drugs, and other stresses, and has anti-inflammatory and immunoregulatory activities. It ameliorates the manifestations of diabetes in preclinical models, suggesting potential applications for the treatment of diabetic patients. This review outlines the actions of GABA relevant to β-cell regeneration, including its signaling mechanisms and potential interactions with other mediators. These studies increase our understanding of the regenerative processes of pancreatic β-cells, and help pave the way for the development of regenerative medicine for diabetes.
Collapse
Affiliation(s)
- Yun Wan
- Department of Endocrinology and Metabolism, Huashan Hospital, Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Medical College, Fudan University, Shanghai, People’s Republic of China
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St Michael’s Hospital, Toronto, ON, Canada
- Departments of Physiology and Medicine, Faculty of Medicine, Toronto, ON, Canada
- Correspondence: Qinghua Wang, Division of Endocrinology and Metabolism, St Michael’s Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada, Tel +1 416 864 6060 ext 77 610, Fax +1 416 864 5140, Email
| | - Gerald J Prud’homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St Michael’s Hospital, Toronto, ON, Canada
| |
Collapse
|
235
|
Hunzicker-Dunn M, Mayo K. Gonadotropin Signaling in the Ovary. KNOBIL AND NEILL'S PHYSIOLOGY OF REPRODUCTION 2015:895-945. [DOI: 10.1016/b978-0-12-397175-3.00020-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
236
|
Ersahin T, Tuncbag N, Cetin-Atalay R. The PI3K/AKT/mTOR interactive pathway. MOLECULAR BIOSYSTEMS 2015; 11:1946-54. [DOI: 10.1039/c5mb00101c] [Citation(s) in RCA: 245] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of the rapamycin (mTOR) signalling pathway is hyperactivated or altered in many cancer types and regulates a broad range of cellular processes including survival, proliferation, growth, metabolism, angiogenesis and metastasis.
Collapse
Affiliation(s)
- Tulin Ersahin
- Cancer Systems Biology Laboratory
- Graduate School of Informatics
- ODTU
- 06800 Ankara
- Turkey
| | - Nurcan Tuncbag
- Cancer Systems Biology Laboratory
- Graduate School of Informatics
- ODTU
- 06800 Ankara
- Turkey
| | - Rengul Cetin-Atalay
- Cancer Systems Biology Laboratory
- Graduate School of Informatics
- ODTU
- 06800 Ankara
- Turkey
| |
Collapse
|
237
|
Hill R, Kalathur RKR, Callejas S, Colaço L, Brandão R, Serelde B, Cebriá A, Blanco-Aparicio C, Pastor J, Futschik M, Dopazo A, Link W. A novel phosphatidylinositol 3-kinase (PI3K) inhibitor directs a potent FOXO-dependent, p53-independent cell cycle arrest phenotype characterized by the differential induction of a subset of FOXO-regulated genes. Breast Cancer Res 2014; 16:482. [PMID: 25488803 PMCID: PMC4303209 DOI: 10.1186/s13058-014-0482-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 11/14/2014] [Indexed: 02/01/2023] Open
Abstract
Introduction The activation of the phosphoinositide 3-kinase (PI3K)/AKT signalling pathway is one the most frequent genetic events in breast cancer, consequently the development of PI3K inhibitors has attracted much attention. Here we evaluate the effect of PI3K inhibition on global gene expression in breast cancer cells. Methods We used a range of methodologies that include in silico compound analysis, in vitro kinase assays, cell invasion assays, proliferation assays, genome-wide transcription studies (Agilent Technologies full genome arrays), gene set enrichment analysis, quantitative real-time PCR, immunoblotting in addition to chromatin immunoprecipitation. Results We defined the physico-chemical and the biological properties of ETP-45658, a novel potent PI3K inhibitor. We demonstrated that ETP-45658 potently inhibited cell proliferation within a broad range of human cancer cells, most potently suppressing the growth of breast cancer cells via inhibiting cell cycle. We show that this response is Forkhead box O (FOXO) protein dependent and p53 independent. Our genome-wide microarray analysis revealed that the cell cycle was the most affected biological process after exposure to ETP-45658 (or our control PI3K inhibitor PI-103), that despite the multiple transcription factors that are regulated by the PI3K/AKT signalling cascade, only the binding sites for FOXO transcription factors were significantly enriched and only a subset of all FOXO-dependent genes were induced. This disparity in gene transcription was not due to differential FOXO promoter recruitment. Conclusions The constitutive activation of PI3Ks and thus the exclusion of FOXO transcription factors from the nucleus is a key feature of breast cancer. Our results presented here highlight that PI3K inhibition activates specific FOXO-dependent genes that mediate cell cycle arrest in breast cancer cells. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0482-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Richard Hill
- IBB-Institute for Biotechnology and Bioengineering, Centre for Molecular and Structural Biomedicine (CBME), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal. .,Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
| | - Ravi Kiran Reddy Kalathur
- IBB-Institute for Biotechnology and Bioengineering, Centre for Molecular and Structural Biomedicine (CBME), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
| | - Sergio Callejas
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle de Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| | - Laura Colaço
- IBB-Institute for Biotechnology and Bioengineering, Centre for Molecular and Structural Biomedicine (CBME), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
| | - Ricardo Brandão
- IBB-Institute for Biotechnology and Bioengineering, Centre for Molecular and Structural Biomedicine (CBME), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
| | - Beatriz Serelde
- Experimental Therapeutics Program, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| | - Antonio Cebriá
- Experimental Therapeutics Program, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| | - Carmen Blanco-Aparicio
- Experimental Therapeutics Program, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| | - Joaquín Pastor
- Experimental Therapeutics Program, Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| | - Matthias Futschik
- IBB-Institute for Biotechnology and Bioengineering, Centre for Molecular and Structural Biomedicine (CBME), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
| | - Ana Dopazo
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle de Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| | - Wolfgang Link
- IBB-Institute for Biotechnology and Bioengineering, Centre for Molecular and Structural Biomedicine (CBME), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal. .,Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
| |
Collapse
|
238
|
Purwana I, Zheng J, Li X, Deurloo M, Son DO, Zhang Z, Liang C, Shen E, Tadkase A, Feng ZP, Li Y, Hasilo C, Paraskevas S, Bortell R, Greiner DL, Atkinson M, Prud'homme GJ, Wang Q. GABA promotes human β-cell proliferation and modulates glucose homeostasis. Diabetes 2014; 63:4197-205. [PMID: 25008178 DOI: 10.2337/db14-0153] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
γ-Aminobutyric acid (GABA) exerts protective and regenerative effects on mouse islet β-cells. However, in humans it is unknown whether it can increase β-cell mass and improve glucose homeostasis. To address this question, we transplanted a suboptimal mass of human islets into immunodeficient NOD-scid-γ mice with streptozotocin-induced diabetes. GABA treatment increased grafted β-cell proliferation, while decreasing apoptosis, leading to enhanced β-cell mass. This was associated with increased circulating human insulin and reduced glucagon levels. Importantly, GABA administration lowered blood glucose levels and improved glucose excursion rates. We investigated GABA receptor expression and signaling mechanisms. In human islets, GABA activated a calcium-dependent signaling pathway through both GABA A receptor and GABA B receptor. This activated the phosphatidylinositol 3-kinase-Akt and CREB-IRS-2 signaling pathways that convey GABA signals responsible for β-cell proliferation and survival. Our findings suggest that GABA regulates human β-cell mass and may be beneficial for the treatment of diabetes or improvement of islet transplantation.
Collapse
Affiliation(s)
- Indri Purwana
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Juan Zheng
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Xiaoming Li
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Marielle Deurloo
- Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Dong Ok Son
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Zhaoyun Zhang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Christie Liang
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Eddie Shen
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Akshaya Tadkase
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Zhong-Ping Feng
- Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Yiming Li
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Craig Hasilo
- Department of Surgery, McGill University, and Human Islet Transplantation Laboratory, McGill University Health Centre, Montreal, Quebec, Canada
| | - Steven Paraskevas
- Department of Surgery, McGill University, and Human Islet Transplantation Laboratory, McGill University Health Centre, Montreal, Quebec, Canada
| | - Rita Bortell
- Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Dale L Greiner
- Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Mark Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Health Science Center, Gainesville, FL
| | - Gerald J Prud'homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Qinghua Wang
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
239
|
Post-injury treatment with 7,8-dihydroxyflavone, a TrkB receptor agonist, protects against experimental traumatic brain injury via PI3K/Akt signaling. PLoS One 2014; 9:e113397. [PMID: 25415296 PMCID: PMC4240709 DOI: 10.1371/journal.pone.0113397] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/23/2014] [Indexed: 01/21/2023] Open
Abstract
Tropomyosin-related kinase B (TrkB) signaling is critical for promoting neuronal survival following brain damage. The present study investigated the effects and underlying mechanisms of TrkB activation by the TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) on traumatic brain injury (TBI). Mice subjected to controlled cortical impact received intraperitoneal 7,8-DHF or vehicle injection 10 min post-injury and subsequently daily for 3 days. Behavioral studies, histology analysis and brain water content assessment were performed. Levels of TrkB signaling-related molecules and apoptosis-related proteins were analyzed. The protective effect of 7,8-DHF was also investigated in primary neurons subjected to stretch injury. Treatment with 20 mg/kg 7,8-DHF attenuated functional deficits and brain damage up to post-injury day 28. 7,8-DHF also reduced brain edema, neuronal death, and apoptosis at day 4. These changes were accompanied by a significant decrease in cleaved caspase-3 and increase in Bcl-2/Bax ratio. 7,8-DHF enhanced phosphorylation of TrkB, Akt (Ser473/Thr308), and Bad at day 4, but had no effect on Erk 1/2 phosphorylation. Moreover, 7,8-DHF increased brain-derived neurotrophic factor levels and promoted cAMP response element-binding protein (CREB) activation. This beneficial effect was attenuated by inhibition of TrkB or PI3K/Akt. 7,8-DHF also promoted survival and reduced apoptosis in cortical neurons subjected to stretch injury. Remarkably, delayed administration of 7,8-DHF at 3 h post-injury reduced brain tissue damage. Our study demonstrates that activation of TrkB signaling by 7,8-DHF protects against TBI via the PI3K/Akt but not Erk pathway, and this protective effect may be amplified via the PI3K/Akt-CREB cascades.
Collapse
|
240
|
Meffre D, Massaad C, Grenier J. Lithium chloride stimulates PLP and MBP expression in oligodendrocytes via Wnt/β-catenin and Akt/CREB pathways. Neuroscience 2014; 284:962-971. [PMID: 25451297 DOI: 10.1016/j.neuroscience.2014.10.064] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 10/27/2014] [Accepted: 10/27/2014] [Indexed: 01/20/2023]
Abstract
Deciphering the molecular pathways involved in myelin gene expression is a major point of interest to better understand re/myelination processes. In this study, we investigated the role of Lithium Chloride (LiCl), a drug largely used for the treatment of neurological disorders, on the two major central myelin gene expression (PLP and MBP) in mouse oligodendrocytes. We show that LiCl enhances the expression of both PLP and MBP, by increasing mRNA amount and promoter activities. We investigated whether Wnt/β-catenin and/or Akt/CREB pathways are modulated by LiCl to regulate myelin gene expression. We showed that β-catenin is required both for PLP and MBP basal promoter activities and for LiCl-induced myelin gene stimulation. Furthermore, while CREB functionality does not influence PLP expression, MBP promoter activity depends on Akt/CREB activation. Finally, we show that LiCl can stimulate oligodendrocyte morphological maturation, and promote remyelination after lysolecithin-induced demyelination of organotypic cerebellar slice cultures. Our data provide mechanistic evidences that Akt/CREB together with β-catenin participate in the transcriptional control of PLP and MBP exerted by LiCl. Therefore, the use of LiCl to balance between β-catenin and CREB effectors could be considered as an efficient remyelinating strategy.
Collapse
Affiliation(s)
- D Meffre
- Paris Descartes University, INSERM UMR-S 1124, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France.
| | - C Massaad
- Paris Descartes University, INSERM UMR-S 1124, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - J Grenier
- Paris Descartes University, INSERM UMR-S 1124, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| |
Collapse
|
241
|
Sathe A, Guerth F, Cronauer MV, Heck MM, Thalgott M, Gschwend JE, Retz M, Nawroth R. Mutant PIK3CA controls DUSP1-dependent ERK 1/2 activity to confer response to AKT target therapy. Br J Cancer 2014; 111:2103-13. [PMID: 25349966 PMCID: PMC4260039 DOI: 10.1038/bjc.2014.534] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/29/2014] [Accepted: 09/08/2014] [Indexed: 11/13/2022] Open
Abstract
Background: Alterations in the phosphoinositide 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) signalling pathway are frequent in urothelial bladder cancer (BLCA) and thus provide a potential target for novel therapeutic strategies. We investigated the efficacy of the AKT inhibitor MK-2206 in BLCA and the molecular determinants that predict therapy response. Methods: Biochemical and functional effects of the AKT inhibitor MK-2206 were analysed on a panel of 11 BLCA cell lines possessing different genetic alterations. Cell viability (CellTiter-Blue, cell counts), apoptosis (caspase 3/7 activity) and cell cycle progression (EdU incorporation) were analysed to determine effects on cell growth and proliferation. cDNA or siRNA transfections were used to manipulate the expression of specific proteins such as wild-type or mutant PIK3CA, DUSP1 or CREB. For in vivo analysis, the chicken chorioallantoic membrane model was utilised and tumours were characterised by weight and biochemically for the expression of Ki-67 and AKT phosphorylation. Results: Treatment with MK-2206 suppressed AKT and S6K1 but not 4E-BP1 phosphorylation in all cell lines. Functionally, only cell lines bearing mutations in the hotspot helical domain of PIK3CA were sensitive to the drug, independent of other genetic alterations in the PI3K or MAPK signalling pathway. Following MK-2206 treatment, the presence of mutant PIK3CA resulted in an increase in DUSP1 expression that induced a decrease in ERK 1/2 phosphorylation. Manipulating the expression of mutant or wild-type PIK3CA or DUSP1 confirmed that this mechanism is responsible for the induction of apoptosis and the inhibition of tumour proliferation in vitro and in vivo, to sensitise cells to AKT target therapy. Conclusion or interpretation: PIK3CA mutations confer sensitivity to AKT target therapy in BLCA by regulating DUSP1 expression and subsequent ERK1/2 dephosphorylation and can potentially serve as a stratifying biomarker for treatment.
Collapse
Affiliation(s)
- A Sathe
- Department of Urology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, Munich 81675, Germany
| | - F Guerth
- Department of Urology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, Munich 81675, Germany
| | - M V Cronauer
- Department of Urology, Ulm University Medical Center, Prittwitzstrasse 43, Ulm 89075, Germany
| | - M M Heck
- Department of Urology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, Munich 81675, Germany
| | - M Thalgott
- Department of Urology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, Munich 81675, Germany
| | - J E Gschwend
- Department of Urology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, Munich 81675, Germany
| | - M Retz
- Department of Urology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, Munich 81675, Germany
| | - R Nawroth
- Department of Urology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, Munich 81675, Germany
| |
Collapse
|
242
|
Shaerzadeh F, Motamedi F, Khodagholi F. Inhibition of akt phosphorylation diminishes mitochondrial biogenesis regulators, tricarboxylic acid cycle activity and exacerbates recognition memory deficit in rat model of Alzheimer's disease. Cell Mol Neurobiol 2014; 34:1223-33. [PMID: 25135709 DOI: 10.1007/s10571-014-0099-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 08/09/2014] [Indexed: 01/01/2023]
Abstract
3-Methyladenine (3-MA), as a PI3K inhibitor, is widely used for inhibition of autophagy. Inhibition of PI3K class I leads to inhibition of Akt phosphorylation, a central molecule involved in diverse arrays of intracellular cascades in nervous system. Accordingly, in the present study, we aimed to determine the alterations of specific mitochondrial biogenesis markers and mitochondrial function in 3-MA-injected rats following amyloid beta (Aβ) insult. Our data revealed that inhibition of Akt phosphorylation downregulates master regulator of mitochondrial biogenesis, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α). Our data also showed that decrease in PGC-1α level presumably is due to decrease in the phosphorylation of cAMP-response element binding and AMP-activated kinase, two upstream activators of PGC-1α. As a consequence, the level of some mitochondrial biogenesis factors including nuclear respiratory factor-1, mitochondrial transcription factor A, and Cytochrome c decreased significantly. Also, activities of tricarboxylic acid cycle (TCA) enzymes such as Aconitase, a-ketoglutarate dehydrogenase, and malate dehydrogenase reduced in the presence of 3-MA with or without Aβ insult. Decrease in mitochondrial biogenesis factors and TCA enzyme activity in the rats receiving 3-MA and Aβ were more compared to the rats that received either alone; indicating the additive destructive effects of these two agents. In agreement with our molecular results, data obtained from behavioral test (using novel objective recognition test) indicated that inhibition of Akt phosphorylation with or without Aβ injection impaired novel recognition (non-spatial) memory. Our results suggest that 3-MA amplified deleterious effects of Aβ by targeting central molecule Akt.
Collapse
Affiliation(s)
- Fatemeh Shaerzadeh
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
243
|
Huang W, Liu X, Cao J, Meng F, Li M, Chen B, Zhang J. miR-134 Regulates Ischemia/Reperfusion Injury-Induced Neuronal Cell Death by Regulating CREB Signaling. J Mol Neurosci 2014; 55:821-9. [DOI: 10.1007/s12031-014-0434-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 09/29/2014] [Indexed: 12/21/2022]
|
244
|
Misuno K, Liu X, Feng S, Hu S. Quantitative proteomic analysis of sphere-forming stem-like oral cancer cells. Stem Cell Res Ther 2014; 4:156. [PMID: 24423398 PMCID: PMC4056689 DOI: 10.1186/scrt386] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 09/16/2013] [Accepted: 11/28/2013] [Indexed: 12/19/2022] Open
Abstract
Introduction The purpose of this study is to identify target proteins that may play important functional roles in oral cancer stem-like cells (CSCs) using mass spectrometry-based quantitative proteomics. Methods Sphere-formation assays were performed on highly invasive UM1 and lowly invasive UM2 oral cancer cell lines, which were derived from the same tongue squamous cell carcinoma, to enrich CSCs. Quantitative proteomic analysis of CSC-like and non-CSC UM1 cells was carried out using tandem mass tagging and two-dimensional liquid chromatography with Orbitrap mass spectrometry. Results CSC-like cancer cells were found to be present in the highly invasive UM1 cell line but absent in the lowly invasive UM2 cell line. Stem cell markers SOX2, OCT4, SOX9 and CD44 were up-regulated, whereas HIF-1 alpha and PGK-1 were down-regulated in CSC-like UM1 cells versus non-CSC UM1 cells. Quantitative proteomic analysis indicated that many proteins in cell cycle, metabolism, G protein signal transduction, translational elongation, development, and RNA splicing pathways were differentially expressed between the two cell phenotypes. Both CREB-1-binding protein (CBP) and phosphorylated CREB-1 were found to be significantly over-expressed in CSC-like UM1 cells. Conclusions CSC-like cells can be enriched from the highly invasive UM1 oral cancer cell line but not from the lowly invasive UM2 oral cancer cell line. There are significant proteomic alterations between CSC-like and non-CSC UM1 cells. In particular, CBP and phosphorylated CREB-1 were significantly up-regulated in CSC-like UM1 cells versus non-CSC UM1 cells, suggesting that the CREB pathway is activated in the CSC-like cells.
Collapse
|
245
|
Rieger DK, Cunha RMS, Lopes MW, Costa AP, Budni J, Rodrigues ALS, Walz R, Teixeira EH, Nascimento KS, Cavada BS, Leal RB. ConBr, a lectin fromCanavalia brasiliensisseeds, modulates signaling pathways and increases BDNF expression probably via a glycosylated target. J Mol Recognit 2014; 27:746-54. [DOI: 10.1002/jmr.2401] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 05/29/2014] [Accepted: 06/01/2014] [Indexed: 01/02/2023]
Affiliation(s)
- Débora K. Rieger
- Departamento de Bioquímica, Centro de Ciências Biológicas; Universidade Federal de Santa Catarina; Florianópolis SC 88040-900 Brazil
| | | | - Mark William Lopes
- Departamento de Bioquímica, Centro de Ciências Biológicas; Universidade Federal de Santa Catarina; Florianópolis SC 88040-900 Brazil
| | - Ana Paula Costa
- Departamento de Bioquímica, Centro de Ciências Biológicas; Universidade Federal de Santa Catarina; Florianópolis SC 88040-900 Brazil
| | - Josiani Budni
- Departamento de Bioquímica, Centro de Ciências Biológicas; Universidade Federal de Santa Catarina; Florianópolis SC 88040-900 Brazil
| | - Ana Lúcia S. Rodrigues
- Departamento de Bioquímica, Centro de Ciências Biológicas; Universidade Federal de Santa Catarina; Florianópolis SC 88040-900 Brazil
| | - Roger Walz
- Departamento de Clínica Médica, Hospital Universitário (HU), Centro de Ciências da Saúde; Universidade Federal de Santa Catarina; Florianópolis SC Brazil
| | - Edson H. Teixeira
- BioMolLab; Universidade Federal do Ceará; Fortaleza CE 60455-970 Brazil
| | | | - Benildo S. Cavada
- BioMolLab; Universidade Federal do Ceará; Fortaleza CE 60455-970 Brazil
| | - Rodrigo B. Leal
- Departamento de Bioquímica, Centro de Ciências Biológicas; Universidade Federal de Santa Catarina; Florianópolis SC 88040-900 Brazil
| |
Collapse
|
246
|
Tian Y, Voineagu I, Paşca SP, Won H, Chandran V, Horvath S, Dolmetsch RE, Geschwind DH. Alteration in basal and depolarization induced transcriptional network in iPSC derived neurons from Timothy syndrome. Genome Med 2014; 6:75. [PMID: 25360157 PMCID: PMC4213483 DOI: 10.1186/s13073-014-0075-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 09/15/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Common genetic variation and rare mutations in genes encoding calcium channel subunits have pleiotropic effects on risk for multiple neuropsychiatric disorders, including autism spectrum disorder (ASD) and schizophrenia. To gain further mechanistic insights by extending previous gene expression data, we constructed co-expression networks in Timothy syndrome (TS), a monogenic condition with high penetrance for ASD, caused by mutations in the L-type calcium channel, Cav1.2. METHODS To identify patient-specific alterations in transcriptome organization, we conducted a genome-wide weighted co-expression network analysis (WGCNA) on neural progenitors and neurons from multiple lines of induced pluripotent stem cells (iPSC) derived from normal and TS (G406R in CACNA1C) individuals. We employed transcription factor binding site enrichment analysis to assess whether TS associated co-expression changes reflect calcium-dependent co-regulation. RESULTS We identified reproducible developmental and activity-dependent gene co-expression modules conserved in patient and control cell lines. By comparing cell lines from case and control subjects, we also identified co-expression modules reflecting distinct aspects of TS, including intellectual disability and ASD-related phenotypes. Moreover, by integrating co-expression with transcription factor binding analysis, we showed the TS-associated transcriptional changes were predicted to be co-regulated by calcium-dependent transcriptional regulators, including NFAT, MEF2, CREB, and FOXO, thus providing a mechanism by which altered Ca(2+) signaling in TS patients leads to the observed molecular dysregulation. CONCLUSIONS We applied WGCNA to construct co-expression networks related to neural development and depolarization in iPSC-derived neural cells from TS and control individuals for the first time. These analyses illustrate how a systems biology approach based on gene networks can yield insights into the molecular mechanisms of neural development and function, and provide clues as to the functional impact of the downstream effects of Ca(2+) signaling dysregulation on transcription.
Collapse
Affiliation(s)
- Yuan Tian
- />Neurogenetics Program, Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
- />Interdepartmental Ph.D. Program in Bioinformatics, University of California, Los Angeles, CA 90095 USA
| | - Irina Voineagu
- />School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052 Australia
| | - Sergiu P Paşca
- />Department of Psychiatry & Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Hyejung Won
- />Neurogenetics Program, Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Vijayendran Chandran
- />Neurogenetics Program, Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Steve Horvath
- />Department of Human Genetics, David Geffen Sch. of Medicine, UCLA, Los Angeles, CA USA
| | - Ricardo E Dolmetsch
- />Department of Neurobiology, Stanford University, Stanford, CA 94305-5345 USA
- />Novartis Institutes for Biomedical Research, Cambridge, MA 02139 USA
| | - Daniel H Geschwind
- />Neurogenetics Program, Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
- />Interdepartmental Ph.D. Program in Bioinformatics, University of California, Los Angeles, CA 90095 USA
- />Department of Human Genetics, David Geffen Sch. of Medicine, UCLA, Los Angeles, CA USA
| |
Collapse
|
247
|
Vu CU, Siddiqui JA, Wadensweiler P, Gayen JR, Avolio E, Bandyopadhyay GK, Biswas N, Chi NW, O'Connor DT, Mahata SK. Nicotinic acetylcholine receptors in glucose homeostasis: the acute hyperglycemic and chronic insulin-sensitive effects of nicotine suggest dual opposing roles of the receptors in male mice. Endocrinology 2014; 155:3793-805. [PMID: 25051446 DOI: 10.1210/en.2014-1320] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cigarette smoking causes insulin resistance. However, nicotine induces anti-inflammation and improves glucose tolerance in insulin-resistant animal models. Here, we determined the effects of nicotine on glucose metabolism in insulin-sensitive C57BL/J6 mice. Acute nicotine administration (30 min) caused fasting hyperglycemia and lowered insulin sensitivity acutely, which depended on the activation of nicotinic-acetylcholine receptors (nAChRs) and correlated with increased catecholamine secretion, nitric oxide (NO) production, and glycogenolysis. Chlorisondamine, an inhibitor of nAChRs, reduced acute nicotine-induced hyperglycemia. qRT-PCR analysis revealed that the liver and muscle express predominantly β4 > α10 > α3 > α7 and β4 > α10 > β1 > α1 mRNA for nAChR subunits respectively, whereas the adrenal gland expresses β4 > α3 > α7 > α10 mRNA. Chronic nicotine treatment significantly suppressed expression of α3-nAChR (predominant peripheral α-subunit) in liver. Whereas acute nicotine treatment raised plasma norepinephrine (NE) and epinephrine (Epi) levels, chronic nicotine exposure raised only Epi. Acute nicotine treatment raised both basal and glucose-stimulated insulin secretion (GSIS). After chronic nicotine treatment, basal insulin level was elevated, but GSIS after acute saline or nicotine treatment was blunted. Chronic nicotine exposure caused an increased buildup of NO in plasma and liver, leading to decreased glycogen storage, along with a concomitant suppression of Pepck and G6Pase mRNA, thus preventing hyperglycemia. The insulin-sensitizing effect of chronic nicotine was independent of weight loss. Chronic nicotine treatment enhanced PI-3-kinase activities and increased Akt and glycogen synthase kinase (GSK)-3β phosphorylation in an nAChR-dependent manner coupled with decreased cAMP response element-binding protein (CREB) phosphorylation. The latter effects caused suppression of Pepck and G6Pase gene expression. Thus, nicotine causes both insulin resistance and insulin sensitivity depending on the duration of the treatment.
Collapse
Affiliation(s)
- Christine U Vu
- VA San Diego Healthcare System (C.U.V., P.W., J.R.G., G.K.B., N.-W.C., D.T.O'C., S.K.M.), San Diego, California 92161; and Department of Medicine (J.A.S., E.A., G.K.B., N.B., N.-W.C., S.K.M.), University of California, San Diego, California 92093
| | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Choi MR, Lee MY, Hong JE, Kim JE, Lee JY, Kim TH, Chun JW, Shin HK, Kim EJ. Rubus coreanus Miquel Ameliorates Scopolamine-Induced Memory Impairments in ICR Mice. J Med Food 2014; 17:1049-56. [DOI: 10.1089/jmf.2013.3004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Mi-Ran Choi
- Center for Efficacy Assessment and Development of Functional Foods and Drugs, Hallym University, Chuncheon, Korea
| | - Min Young Lee
- Center for Efficacy Assessment and Development of Functional Foods and Drugs, Hallym University, Chuncheon, Korea
| | - Ji Eun Hong
- Center for Efficacy Assessment and Development of Functional Foods and Drugs, Hallym University, Chuncheon, Korea
| | - Jeong Eun Kim
- Center for Efficacy Assessment and Development of Functional Foods and Drugs, Hallym University, Chuncheon, Korea
| | - Jae-Yong Lee
- Center for Efficacy Assessment and Development of Functional Foods and Drugs, Hallym University, Chuncheon, Korea
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Korea
| | - Tae Hwan Kim
- Research and Development Center, Hitejinro Co. Ltd., Hongcheon, Korea
| | - Jang Woo Chun
- Research and Development Center, Hitejinro Co. Ltd., Hongcheon, Korea
| | | | - Eun Ji Kim
- Center for Efficacy Assessment and Development of Functional Foods and Drugs, Hallym University, Chuncheon, Korea
| |
Collapse
|
249
|
Ahmad F, Nidadavolu P, Durgadoss L, Ravindranath V. Critical cysteines in Akt1 regulate its activity and proteasomal degradation: implications for neurodegenerative diseases. Free Radic Biol Med 2014; 74:118-28. [PMID: 24933620 DOI: 10.1016/j.freeradbiomed.2014.06.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 06/05/2014] [Accepted: 06/06/2014] [Indexed: 12/25/2022]
Abstract
Impaired Akt1 signaling is observed in neurodegenerative diseases, including Parkinson׳s disease (PD). In PD models oxidative modification of Akt1 leads to its dephosphorylation and consequent loss of its kinase activity. To explore the underlying mechanism we exposed Neuro2A cells to cadmium, a pan inhibitor of protein thiol disulfide oxidoreductases, including glutaredoxin 1 (Grx1), or downregulated Grx1, which led to dephosphorylation of Akt1, loss of its kinase activity, and also decreased Akt1 protein levels. Mutation of cysteines to serines at 296 and 310 in Akt1 did not affect its basal kinase activity but abolished cadmium- and Grx1 downregulation-induced reduction in Akt1 kinase activity, indicating their critical role in redox modulation of Akt1 function and turnover. Cadmium-induced decrease in phosphorylated Akt1 correlated with increased association of wild-type (WT) Akt1 with PP2A, which was absent in the C296-310S Akt1 mutant and was also abolished by N-acetylcysteine treatment. Further, increased proteasomal degradation of Akt1 by cadmium was not seen in the C296-310S Akt1 mutant, indicating that oxidation of cysteine residues facilitates degradation of WT Akt1. Moreover, preventing oxidative modification of Akt1 cysteines 296 and 310 by mutating them to serines increased the cell survival effects of Akt1. Thus, in neurodegenerative states such as PD, maintaining the thiol status of cysteines 296 and 310 in Akt1 would be critical for Akt1 kinase activity and for preventing its degradation by proteasomes. Preventing downregulation of Akt signaling not only has long-range consequences for cell survival but could also affect the multiple roles that Akt plays, including in the Akt-mTOR signaling cascade.
Collapse
Affiliation(s)
- Faraz Ahmad
- Centre for Neuroscience, Indian Institute of Science, Bangalore 560012, India
| | - Prakash Nidadavolu
- Centre for Neuroscience, Indian Institute of Science, Bangalore 560012, India
| | - Lalitha Durgadoss
- Centre for Neuroscience, Indian Institute of Science, Bangalore 560012, India
| | | |
Collapse
|
250
|
Shah KB, Tripathy S, Suganthi H, Rudraiah M. Profiling of luteal transcriptome during prostaglandin F2-alpha treatment in buffalo cows: analysis of signaling pathways associated with luteolysis. PLoS One 2014; 9:e104127. [PMID: 25102061 PMCID: PMC4125180 DOI: 10.1371/journal.pone.0104127] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 07/09/2014] [Indexed: 11/18/2022] Open
Abstract
In several species including the buffalo cow, prostaglandin (PG) F2α is the key molecule responsible for regression of corpus luteum (CL). Experiments were carried out to characterize gene expression changes in the CL tissue at various time points after administration of luteolytic dose of PGF2α in buffalo cows. Circulating progesterone levels decreased within 1 h of PGF2α treatment and evidence of apoptosis was demonstrable at 18 h post treatment. Microarray analysis indicated expression changes in several of immediate early genes and transcription factors within 3 h of treatment. Also, changes in expression of genes associated with cell to cell signaling, cytokine signaling, steroidogenesis, PG synthesis and apoptosis were observed. Analysis of various components of LH/CGR signaling in CL tissues indicated decreased LH/CGR protein expression, pCREB levels and PKA activity post PGF2α treatment. The novel finding of this study is the down regulation of CYP19A1 gene expression accompanied by decrease in expression of E2 receptors and circulating and intra luteal E2 post PGF2α treatment. Mining of microarray data revealed several differentially expressed E2 responsive genes. Since CYP19A1 gene expression is low in the bovine CL, mining of microarray data of PGF2α-treated macaques, the species with high luteal CYP19A1 expression, showed good correlation between differentially expressed E2 responsive genes between both the species. Taken together, the results of this study suggest that PGF2α interferes with luteotrophic signaling, impairs intra-luteal E2 levels and regulates various signaling pathways before the effects on structural luteolysis are manifest.
Collapse
Affiliation(s)
- Kunal B Shah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Sudeshna Tripathy
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Hepziba Suganthi
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Medhamurthy Rudraiah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| |
Collapse
|