1
|
El-ezz DA, Aldahmash W, Esatbeyoglu T, Afifi SM, Elbaset MA. Cilostazol Combats Lipopolysaccharide-Induced Hippocampal Injury in Rats: Role of AKT/GSK3 β/CREB Curbing Neuroinflammation. Adv Pharmacol Pharm Sci 2024; 2024:3465757. [PMID: 39364299 PMCID: PMC11449543 DOI: 10.1155/2024/3465757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 10/05/2024] Open
Abstract
Neuroinflammation is important in the pathophysiology of several degenerative brain disorders. This study looked at the potential neuroprotective benefits of cilostazol, a phosphodiesterase inhibitor, against LPS-induced hippocampus damage in rodents and the principal molecular involvement of AKT/GSK3β/CREB signaling pathways. Behavioral tests revealed that cilostazol successfully corrected LPS-induced neurobehavioral impairments. Furthermore, cilostazol therapy lowered hippocampal levels of amyloid beta 1-42 (Aβ1-42) and p-tau protein, both of which are critical pathological indicators of neurodegenerative disorders. Furthermore, cilostazol administration suppressed LPS-induced rises in hippocampus caspase-3 and NF-κB levels while elevating rat B-cell/lymphoma 2 (BCL2) and brain-derived neurotrophic factor (BDNF) levels, which are implicated in neuronal survival and synaptic plasticity. Cilostazol treatment also restored the decreased phosphorylation of protein kinase B (p-AKT) and reduced the elevated levels of phosphorylated glycogen synthase kinase-3 beta (p-GSK3β) and cAMP response element-binding protein (CREB) in the hippocampus of LPS-treated rats. Histopathological examination revealed that cilostazol ameliorated LPS-induced brain damage with reduced neuronal loss and gliosis. Immunohistochemistry analysis showed a decrease in Iba-1 expression, indicating a reduction in microglial activation in the cilostazol-treated group compared to the LPS group. The findings advocate that cilostazol exerts neuroprotective effects against LPS-induced hippocampal injury by modulating the AKT/GSK3β/CREB pathway and curbing neuroinflammation. Cilostazol may hold promise as a therapeutic agent for neuroinflammatory conditions associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Doaa Abou El-ezz
- Pharmacology and Toxicology DepartmentFaculty of PharmacyOctober University for Modern Sciences and Arts University, Giza 12556, Egypt
| | - Waleed Aldahmash
- Department of ZoologyCollege of ScienceKing Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Tuba Esatbeyoglu
- Department of Molecular Food Chemistry and Food DevelopmentInstitute of Food and One HealthGottfried Wilhelm Leibniz University Hannover, Am Kleinen Felde 30, Hannover 30167, Germany
| | - Sherif M. Afifi
- Department for Life Quality StudiesRimini CampusUniversity of Bologna, Corso d'Augusto 237, Rimini 47921, Italy
| | - Marawan Abd Elbaset
- Department of PharmacologyMedical Research and Clinical Studies InstituteNational Research Centre, Dokki, Giza 12622, Egypt
| |
Collapse
|
2
|
Guo F, Yao L, Zhang W, Chen P, Hao R, Huang X, Jiang J, Wu S. The therapeutic mechanism of Yuye decoction on type 2 diabetes mellitus based on network pharmacology and experimental verification. JOURNAL OF ETHNOPHARMACOLOGY 2023; 308:116222. [PMID: 36828194 DOI: 10.1016/j.jep.2023.116222] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/21/2023] [Accepted: 01/28/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yuye decoction (YYD) has been widely used as a folk Chinese herbal formula in clinical treatment of type 2 diabetes mellitus(T2DM) for many years. However, its mechanism is still unclear. AIM OF THE STUDY The aim of this study was to explore the potential mechanism of YYD against T2DM initially by UHPLC-MS/MS combining with network pharmacology, molecular docking techniques and experimental validation. MATERIALS AND METHODS The main ingredients in the water extract of YYD were initially identified using UHPLC-MS/MS analysis. Combined with network pharmacology and molecular docking techniques, the YYD key compounds-core targets-key signaling pathways network was constructed and the binding activity of key components to core targets was validated. The T2DM rat model was induced by Streptozotocin combined with high glucose and high fat diets. The apoptosis cell model of mouse islet β-cell of Min6 was induced by high-glucose and palmitic acid. Histopathological and immunofluorescence satining were used to evaluate pancreatic islet β-cell function and apoptosis in rats. Min6 cell viability and apoptosis ratio were evaluated by CCK-8 and TUNEL staining. The predicted targets and pathways were validated by experiments in vitro and in vivo. RESULTS The 56 compounds from YYD were identified by UHPLC-MS/MS. The potential targets of the above compounds were predicted by online compound target database, among of which 362 targets were associated with T2DM. Protein-protein interaction analysis identified the main targets such as SRC, MAPK1, PIK3R1, AKT1, HRAS and HSP90AA1, which were considered as the therapeutic targets of YYD on against T2DM. Functional enrichment analysis revealed that PI3K/AKT, FoxO and apoptosis signaling pathways were significantly enriched. Molecular docking results showed that compounds of monolinolein, neomangiferin, mangiferin, pelargonidin-3-O-glucoside and acacetin from YYD had high binding activities to PIK3R1, AKT1, Sirt1 and FoxO1. Therefore, PI3K/AKT1, Sirt1/FoxO1 and apoptotic signaling pathways were considered as predicted targets for experimental validation study. Animal experiments showed that YYD reduced blood glucose levels, improved pancreatic dysfunction and pancreatic islet β-cells apoptosis in T2DM rats which contributed to the activation of AKT1 and FoxO1 and their related signaling molecules. These results were confirmed in Min6 cell model induced by high-glucose and palmitic acid. CONCLUSIONS In summary, this study systematically visualized the possible therapeutic effects and mechanisms of YYD on T2DM through the network pharmacology approach and experimental study. The results indicated that YYD could prevent pancreatic islet dysfunction and reverse islet of β-cells apoptosis possibly via PI3K/AKT1, Sirt1/FoxO1 signaling pathways.
Collapse
Affiliation(s)
- Feng Guo
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, 830017, China
| | - Lan Yao
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, 830017, China.
| | - Wenxiang Zhang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, 830017, China
| | - Pengde Chen
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, 830017, China
| | - Rui Hao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Xuelian Huang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Jie Jiang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, 830017, China
| | - Siyu Wu
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, 830017, China
| |
Collapse
|
3
|
Abstract
Circadian clocks are biochemical time-keeping machines that synchronize animal behavior and physiology with planetary rhythms. In Drosophila, the core components of the clock comprise a transcription/translation feedback loop and are expressed in seven neuronal clusters in the brain. Although it is increasingly evident that the clocks in each of the neuronal clusters are regulated differently, how these clocks communicate with each other across the circadian neuronal network is less clear. Here, we review the latest evidence that describes the physical connectivity of the circadian neuronal network . Using small ventral lateral neurons as a starting point, we summarize how one clock may communicate with another, highlighting the signaling pathways that are both upstream and downstream of these clocks. We propose that additional efforts are required to understand how temporal information generated in each circadian neuron is integrated across a neuronal circuit to regulate rhythmic behavior.
Collapse
Affiliation(s)
- Myra Ahmad
- Department of Pediatrics, Division of Medical Genetics, Dalhousie University, Halifax, NS, Canada
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Wanhe Li
- Laboratory of Genetics, The Rockefeller University, New York, NY, USA
| | - Deniz Top
- Department of Pediatrics, Division of Medical Genetics, Dalhousie University, Halifax, NS, Canada
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
4
|
Liu T, Song Y, Hu A. Neuroprotective mechanisms of mangiferin in neurodegenerative diseases. Drug Dev Res 2021; 82:494-502. [PMID: 33458836 DOI: 10.1002/ddr.21783] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 11/10/2022]
Abstract
The central nervous system (CNS) regulates and coordinates an extensive array of complex processes requiring harmonious regulation of specific genes. CNS disorders represent a large burden on society and cause enormous disability and economic losses. Traditional Chinese medicine (TCM) has been used for many years in the treatment of neurological illnesses, such as Alzheimer's disease, Parkinson's disease, stroke, and depression, as the combination of TCM and Western medicine has superior therapeutic efficacy and minimal toxic side effects. Mangiferin (MGF) is an active compound of the traditional Chinese herb rhizome anemarrhenae, which has antioxidant, anti-inflammation, anti-lipid peroxidation, immunomodulatory, and anti-apoptotic functions in the CNS. MGF has been demonstrated to have therapeutic effects in CNS diseases through a multitude of mechanisms. This review outlines the latest research on the neuroprotective ability of MGF and the diverse molecular mechanisms involved.
Collapse
Affiliation(s)
- Tingjun Liu
- Center of Animal Laboratory, Xuzhou Medical University, Xuzhou, PR China
| | - Yuanjian Song
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China.,Department of Genetics, Xuzhou Engineering Research Center of Medical Genetics and Transformation, Xuzhou Medical University, Xuzhou, China
| | - Ankang Hu
- Center of Animal Laboratory, Xuzhou Medical University, Xuzhou, PR China
| |
Collapse
|
5
|
Role of Microglia in Modulating Adult Neurogenesis in Health and Neurodegeneration. Int J Mol Sci 2020; 21:ijms21186875. [PMID: 32961703 PMCID: PMC7555074 DOI: 10.3390/ijms21186875] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023] Open
Abstract
Microglia are the resident immune cells of the brain, constituting the powerhouse of brain innate immunity. They originate from hematopoietic precursors that infiltrate the developing brain during different stages of embryogenesis, acquiring a phenotype characterized by the presence of dense ramifications. Microglial cells play key roles in maintaining brain homeostasis and regulating brain immune responses. They continuously scan and sense the brain environment to detect any occurring changes. Upon detection of a signal related to physiological or pathological processes, the cells are activated and transform to an amoeboid-like phenotype, mounting adequate responses that range from phagocytosis to secretion of inflammatory and trophic factors. The overwhelming evidence suggests that microglia are crucially implicated in influencing neuronal proliferation and differentiation, as well as synaptic connections, and thereby cognitive and behavioral functions. Here, we review the role of microglia in adult neurogenesis under physiological conditions, and how this role is affected in neurodegenerative diseases.
Collapse
|
6
|
Wang D, Zhao J, Li S, Wei J, Nan L, Mallampalli RK, Weathington NM, Ma H, Zhao Y. Phosphorylated E2F1 is stabilized by nuclear USP11 to drive Peg10 gene expression and activate lung epithelial cells. J Mol Cell Biol 2019; 10:60-73. [PMID: 28992046 DOI: 10.1093/jmcb/mjx034] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 08/15/2017] [Indexed: 12/21/2022] Open
Abstract
Phosphorylation affects ubiquitination, stability, and activity of transcriptional factors, thus regulating various cellular functions. E2F transcriptional factor 1 (E2F1) regulates paternally expressed imprinted gene 10 (Peg10) expression, thereby promoting cell proliferation. However, the effect of E2F1 stability on Peg10 expression and the molecular regulation of E2F1 stability by its phosphorylation have not been well demonstrated. Here, we describe a new pathway in which phosphorylation of E2F1 by GSK3β increases E2F1 association with the deubiquitinating enzyme, ubiquitin-specific protease 11 (USP11), which removes K63-linked ubiquitin chains thereby preventing E2F1 degradation in the nuclei. Downregulation of USP11 increases E2F1 ubiquitination and reduces E2F1 stability and protein levels, thereby decreasing Peg10 mRNA levels. Physiologically, USP11 depletion suppresses cell proliferation and wound healing in lung epithelial cells, and these effects are reversed by E2F1 and PEG10 overexpression. Thus, our study reveals a new molecular model that phosphorylation promotes substrate stability through increasing its association with a deubiquitinating enzyme. The data suggest that GSK3β and USP11 act in concert to modulate E2F1 abundance and PEG10 expression in lung epithelial cells to affect cell wound healing. This study provides new therapeutic targets to lessen lung injury by improving lung epithelial cell repair and remodeling after injury.
Collapse
Affiliation(s)
- Dan Wang
- Department of Anesthesia, the First Affiliated Hospital of Jilin University, Changchun, China
| | - Jing Zhao
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Shuang Li
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, USA.,Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jianxin Wei
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Ling Nan
- Department of Anesthesia, the First Affiliated Hospital of Jilin University, Changchun, China
| | - Rama K Mallampalli
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, USA.,Acute Lung Injury Center of Excellence and Vascular Medical Institute, University of Pittsburgh, Pittsburgh, USA
| | - Nathaniel M Weathington
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, USA.,Acute Lung Injury Center of Excellence and Vascular Medical Institute, University of Pittsburgh, Pittsburgh, USA
| | - Haichun Ma
- Department of Anesthesia, the First Affiliated Hospital of Jilin University, Changchun, China
| | - Yutong Zhao
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, USA.,Acute Lung Injury Center of Excellence and Vascular Medical Institute, University of Pittsburgh, Pittsburgh, USA
| |
Collapse
|
7
|
Rahman MK, You M. Molecular cloning and transcriptional and functional analysis of glycogen synthase kinase-3β in Haemaphysalis longicornis (Acari, Ixodidae). ACTA ACUST UNITED AC 2019; 26:39. [PMID: 31294687 PMCID: PMC6621911 DOI: 10.1051/parasite/2019038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/17/2019] [Indexed: 12/29/2022]
Abstract
Glycogen synthase kinase 3 (GSK-3), which belongs to the serine/threonine kinase family, regulates glycogen metabolism, Wnt signaling, hormonal regulation, and embryonic development in many eukaryotes. Here, we cloned a complete open reading frame (ORF) of glycogen synthase kinase 3β (GSK-3β) from Haemaphysalis longicornis and characterized its transcriptional and functional status. The ORF of GSK-3β possesses 1242 nucleotides encoding a mature protein of 413 amino acid residues. GSK-3β nucleotide and protein sequences are highly conserved among different vertebrate and invertebrate animals, with identity between 47.8–100% and 63.2–88.7%, respectively. Sequence comparison showed one signature domain between the residues of 51 and 335 amino acids, which was identified as a protein kinase (serine/threonine). RT-PCR showed GSK-3β mRNA present in all developmental stages of H. longicornis. Interestingly, a higher transcript level was observed in nymph and 7-day-old eggs compared with others by real-time PCR, indicating a role of GSK-3β in the early stages of life. The functional status of GSK-3β was characterized by RNA interference (RNAi) and caused significant (p < 0.05) reduction in feeding and reproduction, as well as an abnormality in eggs and hatching. Taken together, our results suggest that GSK-3β may be an important candidate for a multiple antigen vaccine for controlling the tick population.
Collapse
Affiliation(s)
- Md Khalesur Rahman
- Laboratory of Veterinary Parasitology, College of Veterinary Medicine and Bio-Safety Research Centre, Chonbuk National University, Iksan 54596, Republic of Korea - Department of Microbiology, Faculty of Veterinary and Animal Science, Hajee Mohammad Danesh Science and Technology University, Dinajpur 5200, Bangladesh
| | - Myungjo You
- Laboratory of Veterinary Parasitology, College of Veterinary Medicine and Bio-Safety Research Centre, Chonbuk National University, Iksan 54596, Republic of Korea
| |
Collapse
|
8
|
Saxena A, Scaini G, Bavaresco DV, Leite C, Valvassori SS, Carvalho AF, Quevedo J. Role of Protein Kinase C in Bipolar Disorder: A Review of the Current Literature. MOLECULAR NEUROPSYCHIATRY 2017; 3:108-124. [PMID: 29230399 DOI: 10.1159/000480349] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/14/2017] [Indexed: 12/19/2022]
Abstract
Bipolar disorder (BD) is a major health problem. It causes significant morbidity and imposes a burden on the society. Available treatments help a substantial proportion of patients but are not beneficial for an estimated 40-50%. Thus, there is a great need to further our understanding the pathophysiology of BD to identify new therapeutic avenues. The preponderance of evidence pointed towards a role of protein kinase C (PKC) in BD. We reviewed the literature pertinent to the role of PKC in BD. We present recent advances from preclinical and clinical studies that further support the role of PKC. Moreover, we discuss the role of PKC on synaptogenesis and neuroplasticity in the context of BD. The recent development of animal models of BD, such as stimulant-treated and paradoxical sleep deprivation, and the ability to intervene pharmacologically provide further insights into the involvement of PKC in BD. In addition, the effect of PKC inhibitors, such as tamoxifen, in the resolution of manic symptoms in patients with BD further points in that direction. Furthermore, a wide variety of growth factors influence neurotransmission through several molecular pathways that involve downstream effects of PKC. Our current understanding identifies the PKC pathway as a potential therapeutic avenue for BD.
Collapse
Affiliation(s)
- Ashwini Saxena
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Giselli Scaini
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Daniela V Bavaresco
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, Brazil
| | - Camila Leite
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, Brazil
| | - Samira S Valvassori
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, Brazil
| | - André F Carvalho
- Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceara, Fortaleza, Brazil
| | - João Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, Brazil.,Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, USA
| |
Collapse
|
9
|
AKT/GSK3β signaling pathway is critically involved in human pluripotent stem cell survival. Sci Rep 2016; 6:35660. [PMID: 27762303 PMCID: PMC5071844 DOI: 10.1038/srep35660] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 10/04/2016] [Indexed: 01/24/2023] Open
Abstract
Human embryonic and induced pluripotent stem cells are self-renewing pluripotent stem cells (PSC) that can differentiate into a wide range of specialized cells. Basic fibroblast growth factor is essential for PSC survival, stemness and self-renewal. PI3K/AKT pathway regulates cell viability and apoptosis in many cell types. Although it has been demonstrated that PI3K/AKT activation by bFGF is relevant for PSC stemness maintenance its role on PSC survival remains elusive. In this study we explored the molecular mechanisms involved in the regulation of PSC survival by AKT. We found that inhibition of AKT with three non-structurally related inhibitors (GSK690693, AKT inhibitor VIII and AKT inhibitor IV) decreased cell viability and induced apoptosis. We observed a rapid increase in phosphatidylserine translocation and in the extent of DNA fragmentation after inhibitors addition. Moreover, abrogation of AKT activity led to Caspase-9, Caspase-3, and PARP cleavage. Importantly, we demonstrated by pharmacological inhibition and siRNA knockdown that GSK3β signaling is responsible, at least in part, of the apoptosis triggered by AKT inhibition. Moreover, GSK3β inhibition decreases basal apoptosis rate and promotes PSC proliferation. In conclusion, we demonstrated that AKT activation prevents apoptosis, partly through inhibition of GSK3β, and thus results relevant for PSC survival.
Collapse
|
10
|
Glycogen Synthase Kinase-3 is involved in glycogen metabolism control and embryogenesis of Rhodnius prolixus. Parasitology 2016; 143:1569-79. [DOI: 10.1017/s0031182016001487] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SUMMARYRhodnius prolixus is a blood-feeding insect that transmits Trypanosoma cruzi and Trypanosoma rangeli to vertebrate hosts. Rhodnius prolixus is also a classical model in insect physiology, and the recent availability of R. prolixus genome has opened new avenues on triatomine research. Glycogen synthase kinase 3 (GSK-3) is classically described as a key enzyme involved in glycogen metabolism, also acting as a downstream component of the Wnt pathway during embryogenesis. GSK-3 has been shown to be highly conserved among several organisms, mainly in the catalytic domain region. Meanwhile, the role of GSK-3 during R. prolixus embryogenesis or glycogen metabolism has not been investigated. Here we show that chemical inhibition of GSK-3 by alsterpaullone, an ATP-competitive inhibitor of GSK3, does not affect adult survival rate, though it alters oviposition and egg hatching. Specific GSK-3 gene silencing by dsRNA injection in adult females showed a similar phenotype. Furthermore, bright field and 4’−6-diamidino-2-phenylindole (DAPI) staining analysis revealed that ovaries and eggs from dsGSK-3 injected females exhibited specific morphological defects. We also demonstrate that glycogen content was inversely related to activity and transcription levels of GSK-3 during embryogenesis. Lastly, after GSK-3 knockdown, we observed changes in the expression of the Wingless (Wnt) downstream target β-catenin as well as in members of other pathways such as the receptor Notch. Taken together, our results show that GSK-3 regulation is essential for R. prolixus oogenesis and embryogenesis.
Collapse
|
11
|
Wang Y, Tong K. Glycogen synthase kinase-3β inhibitor ameliorates imbalance of connexin 43 in an acute kidney injury model. Toxicol Rep 2015; 2:1391-1395. [PMID: 28962480 PMCID: PMC5598357 DOI: 10.1016/j.toxrep.2015.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 10/10/2015] [Accepted: 10/11/2015] [Indexed: 01/14/2023] Open
Abstract
This study was designed to evaluate whether glycogen synthase kinase-3β (GSK-3β) inhibitor, 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8) induced the the expression of connexin 43 (Cx43) to protect against renal ischemia–reperfusion (I/R) injury (RI/RI) in rats. Rats were subjected to 45 min ischemia followed 2 h reperfusion with TDZD-8 (1 mg/kg) for 5 min prior to reperfusion. The results indicated that TDZD-8 improved the recovery of renal function, reduced oxidative stress and inflammation injury, and upregulated the expression of (Cx43) as compared to I/R group. Therefore, our study demonstrated that TDZD-8 provided a protection to the kidney against I/R injury in rats through inducing the expression of (Cx43).
Collapse
Affiliation(s)
- Yini Wang
- Department of Nursing, Medical College, Shantou University, 26 Xinling Road, Shantou 515041, PR China.,Southwest University of Science and Technology, College of Life Science and Engineering, Mianyang, Sichuan 621010, PR China
| | - Ke Tong
- Department of Nursing, Medical College, Shantou University, 26 Xinling Road, Shantou 515041, PR China.,Southwest University of Science and Technology, College of Life Science and Engineering, Mianyang, Sichuan 621010, PR China
| |
Collapse
|
12
|
Paeng J, Chang JH, Lee SH, Nam BY, Kang HY, Kim S, Oh HJ, Park JT, Han SH, Yoo TH, Kang SW. Enhanced glycogen synthase kinase-3β activity mediates podocyte apoptosis under diabetic conditions. Apoptosis 2014; 19:1678-90. [DOI: 10.1007/s10495-014-1037-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
13
|
Silvestri I, Testa F, Zappasodi R, Cairo CW, Zhang Y, Lupo B, Galli R, Di Nicola M, Venerando B, Tringali C. Sialidase NEU4 is involved in glioblastoma stem cell survival. Cell Death Dis 2014; 5:e1381. [PMID: 25144716 PMCID: PMC4454322 DOI: 10.1038/cddis.2014.349] [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] [Received: 04/15/2014] [Revised: 07/08/2014] [Accepted: 07/09/2014] [Indexed: 01/30/2023]
Abstract
The human sialidase, NEU4, has emerged as a possible regulator of neuronal differentiation and its overexpression has been demonstrated to promote the acquisition of a stem cell-like phenotype in neuroblastoma cells. In this paper, we demonstrated that glioblastoma stem cells (GSCs) isolated from glioblastoma multiforme (GBM) cell lines and patients' specimens as neurospheres are specifically marked by the upregulation of NEU4; in contrast, the expression of NEU4 is very low in non-neurosphere-differentiated GBM cells. We showed that NEU4 silencing by miRNA or a chemical inhibitor of its catalytic activity triggered key events in GSCs, including (a) the activation of the glycogen synthase kinase 3β, with the consequent inhibition of Sonic Hedgehog and Wnt/β-catenin signalling pathways; (b) the decrease of the stem cell-like gene expression and marker signatures, evidenced by the reduction of NANOG, OCT-4, SOX-2, CD133 expression, ganglioside GD3 synthesis, and an altered protein glycosylation profile; and (c) a significant decrease in GSCs survival. Consistent with this finding, increased NEU4 activity and expression induced in the more differentiated GBM cells by the NEU4 agonist thymoquinone increased the expression of OCT-4 and GLI-1. Thus, NEU4 expression and activity appeared to help to determine the molecular signature of GSCs and to be closely connected with their survival properties. Given the pivotal role played by GSCs in GBM lethality, our results strongly suggest that NEU4 inhibition could significantly improve current therapies against this tumour.
Collapse
Affiliation(s)
- I Silvestri
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate (Milan), Italy
| | - F Testa
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate (Milan), Italy
| | - R Zappasodi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | - C W Cairo
- Alberta Glycomics Center, Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Y Zhang
- Alberta Glycomics Center, Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - B Lupo
- Laboratory of Molecular Pharmacology, Institute for Cancer Research and Treatment (IRCC), Candiolo (Torino), Italy
| | - R Galli
- Neural Stem Cell Biology Unit, Division of Regenerative Medicine Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - M Di Nicola
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | - B Venerando
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate (Milan), Italy
| | - C Tringali
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate (Milan), Italy
| |
Collapse
|
14
|
da Rocha Fernandes M, Martins R, Pessoa Costa E, Casagrande Pacidônio E, Araujo de Abreu L, da Silva Vaz I, Moreira LA, da Fonseca RN, Logullo C. The modulation of the symbiont/host interaction between Wolbachia pipientis and Aedes fluviatilis embryos by glycogen metabolism. PLoS One 2014; 9:e98966. [PMID: 24926801 PMCID: PMC4057193 DOI: 10.1371/journal.pone.0098966] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 05/08/2014] [Indexed: 12/22/2022] Open
Abstract
Wolbachia pipientis, a maternally transmitted bacterium that colonizes arthropods, may affect the general aspects of insect physiology, particularly reproduction. Wolbachia is a natural endosymbiont of Aedes fluviatilis, whose effects in embryogenesis and reproduction have not been addressed so far. In this context, we investigated the correlation between glucose metabolism and morphological alterations during A. fluviatilis embryo development in Wolbachia-positive (W+) and Wolbachia-negative (W-) mosquito strains. While both strains do not display significant morphological and larval hatching differences, larger differences were observed in hexokinase activity and glycogen contents during early and mid-stages of embryogenesis, respectively. To investigate if glycogen would be required for parasite-host interaction, we reduced Glycogen Synthase Kinase-3 (GSK-3) levels in adult females and their eggs by RNAi. GSK-3 knock-down leads to embryonic lethality, lower levels of glycogen and total protein and Wolbachia reduction. Therefore, our results suggest that the relationship between A. fluviatilis and Wolbachia may be modulated by glycogen metabolism.
Collapse
Affiliation(s)
- Mariana da Rocha Fernandes
- Laboratório de Sanidade Animal, Laboratório de Química e Função de Proteínas e Peptídeos and Unidade de Experimentação Animal - RJ, Universidade Estadual do Norte Fluminense Darcy Ribeiro (UENF), Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Renato Martins
- Laboratório de Sanidade Animal, Laboratório de Química e Função de Proteínas e Peptídeos and Unidade de Experimentação Animal - RJ, Universidade Estadual do Norte Fluminense Darcy Ribeiro (UENF), Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Evenilton Pessoa Costa
- Laboratório de Sanidade Animal, Laboratório de Química e Função de Proteínas e Peptídeos and Unidade de Experimentação Animal - RJ, Universidade Estadual do Norte Fluminense Darcy Ribeiro (UENF), Campos dos Goytacazes, Rio de Janeiro, Brazil
| | | | - Leonardo Araujo de Abreu
- Laboratório de Sanidade Animal, Laboratório de Química e Função de Proteínas e Peptídeos and Unidade de Experimentação Animal - RJ, Universidade Estadual do Norte Fluminense Darcy Ribeiro (UENF), Campos dos Goytacazes, Rio de Janeiro, Brazil
- Laboratório Integrado de Bioquímica Hatisaburo Masuda (LIBHM), Núcleo de Pesquisas Ecológicas e Socioambientais de Macaé (NUPEM), Universidade Federal do Rio de Janeiro (UFRJ/Macaé), Rio de Janeiro, Brazil
| | - Itabajara da Silva Vaz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Rio Grande do Sul, Brazil
| | - Luciano A. Moreira
- Laboratório de Malária, Centro de Pesquisas René Rachou - Fiocruz, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo Nunes da Fonseca
- Laboratório Integrado de Bioquímica Hatisaburo Masuda (LIBHM), Núcleo de Pesquisas Ecológicas e Socioambientais de Macaé (NUPEM), Universidade Federal do Rio de Janeiro (UFRJ/Macaé), Rio de Janeiro, Brazil
| | - Carlos Logullo
- Laboratório de Sanidade Animal, Laboratório de Química e Função de Proteínas e Peptídeos and Unidade de Experimentação Animal - RJ, Universidade Estadual do Norte Fluminense Darcy Ribeiro (UENF), Campos dos Goytacazes, Rio de Janeiro, Brazil
| |
Collapse
|
15
|
Abstract
Increased endothelial permeability and reduction of alveolar liquid clearance capacity are two leading pathogenic mechanisms of pulmonary edema, which is a major complication of acute lung injury, severe pneumonia, and acute respiratory distress syndrome, the pathologies characterized by unacceptably high rates of morbidity and mortality. Besides the success in protective ventilation strategies, no efficient pharmacological approaches exist to treat this devastating condition. Understanding of fundamental mechanisms involved in regulation of endothelial permeability is essential for development of barrier protective therapeutic strategies. Ongoing studies characterized specific barrier protective mechanisms and identified intracellular targets directly involved in regulation of endothelial permeability. Growing evidence suggests that, although each protective agonist triggers a unique pattern of signaling pathways, selected common mechanisms contributing to endothelial barrier protection may be shared by different barrier protective agents. Therefore, understanding of basic barrier protective mechanisms in pulmonary endothelium is essential for selection of optimal treatment of pulmonary edema of different etiology. This article focuses on mechanisms of lung vascular permeability, reviews major intracellular signaling cascades involved in endothelial monolayer barrier preservation and summarizes a current knowledge regarding recently identified compounds which either reduce pulmonary endothelial barrier disruption and hyperpermeability, or reverse preexisting lung vascular barrier compromise induced by pathologic insults.
Collapse
Affiliation(s)
- Konstantin G Birukov
- Lung Injury Center, Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois, USA.
| | | | | |
Collapse
|
16
|
Matsuzaki E, Hiratsuka S, Hamachi T, Takahashi-Yanaga F, Hashimoto Y, Higashi K, Kobayashi M, Hirofuji T, Hirata M, Maeda K. Sphingosine-1-phosphate promotes the nuclear translocation of β-catenin and thereby induces osteoprotegerin gene expression in osteoblast-like cell lines. Bone 2013; 55:315-24. [PMID: 23612487 DOI: 10.1016/j.bone.2013.04.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 04/05/2013] [Accepted: 04/15/2013] [Indexed: 11/20/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a well-known signaling sphingolipid and bioactive lipid mediator. Recently, it was reported that S1P inhibits osteoclast differentiation and bone resorption. On the other hand, S1P effects on osteoblasts and bone formation are little known. In this study, we investigated the effects of S1P on osteoblasts, using two osteoblast-like cell lines, SaOS-2 and MC3T3-E1. S1P activated phosphatidylinositol 3-kinase (PI3K)/Akt signaling, leading to the inhibition of glycogen synthase kinase-3β and the nuclear translocation of β-catenin, followed by the increase of the transcriptional activity by β-catenin/T-cell factor complex formation in both SaOS-2 cells and MC3T3-E1 cells. The inhibitors of PI3K and Akt suppressed S1P-induced nuclear localization of β-catenin. We further investigated the effects of PI3K/Akt signaling on the Wnt/β-catenin signaling pathway, since β-catenin takes a central role in this signaling pathway. Both inhibitors for PI3K and Akt suppressed the nuclear localization of β-catenin and T-cell factor transcriptional activity induced by Wnt-3a. S1P increased the amount of osteoprotegerin at both mRNA and protein levels, and increased the activity of alkaline phosphatase, leading to the mineralization. These findings suggest that S1P activates the PI3K/Akt signaling pathway leading to the promotion of nuclear translocation of β-catenin in osteoblast-like cells, resulting in the upregulation of osteoptotegerin and osteoblast differentiation markers including alkaline phosphatase, probably relating to the inhibition of osteoclast formation and the mineralization, respectively.
Collapse
Affiliation(s)
- Etsuko Matsuzaki
- Periodontal Section, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Pal I, Mandal M. PI3K and Akt as molecular targets for cancer therapy: current clinical outcomes. Acta Pharmacol Sin 2012; 33:1441-58. [PMID: 22983389 DOI: 10.1038/aps.2012.72] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The PI3K-Akt pathway is a vital regulator of cell proliferation and survival. Alterations in the PIK3CA gene that lead to enhanced PI3K kinase activity have been reported in many human cancer types, including cancers of the colon, breast, brain, liver, stomach and lung. Deregulation of PI3K causes aberrant Akt activity. Therefore targeting this pathway could have implications for cancer treatment. The first generation PI3K-Akt inhibitors were proven to be highly effective with a low IC(50), but later, they were shown to have toxic side effects and poor pharmacological properties and selectivity. Thus, these inhibitors were only effective in preclinical models. However, derivatives of these first generation inhibitors are much more selective and are quite effective in targeting the PI3K-Akt pathway, either alone or in combination. These second-generation inhibitors are essentially a specific chemical moiety that helps to form a strong hydrogen bond interaction with the PI3K/Akt molecule. The goal of this review is to delineate the current efforts that have been undertaken to inhibit the various components of the PI3K and Akt pathway in different types of cancer both in vitro and in vivo. Our focus here is on these novel therapies and their inhibitory effects that depend upon their chemical nature, as well as their development towards clinical trials.
Collapse
|
18
|
Fairbairn EA, Bonthius J, Cherr GN. Polycyclic aromatic hydrocarbons and dibutyl phthalate disrupt dorsal-ventral axis determination via the Wnt/β-catenin signaling pathway in zebrafish embryos. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2012; 124-125:188-196. [PMID: 22975441 DOI: 10.1016/j.aquatox.2012.08.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/20/2012] [Accepted: 08/21/2012] [Indexed: 06/01/2023]
Abstract
The canonical Wnt/β-catenin signaling pathway is critical during early teleost development for establishing the dorsal-ventral axis. Within this pathway, GSK-3β, a key regulatory kinase in the Wnt pathway, regulates β-catenin degradation and thus the ability of β-catenin to enter nuclei, where it can activate expression of genes that have been linked to the specification of the dorsal-ventral axis. In this study, we describe the morphological abnormalities that resulted in zebrafish embryos when axis determination was disrupted by environmental contaminants. These abnormalities were linked to abnormal nuclear accumulation of β-catenin. Furthermore, we demonstrated that the developmental abnormalities and altered nuclear β-catenin accumulation occurred when embryos were exposed to commercial GSK-3β inhibitors. Zebrafish embryos were exposed to commercially available GSK-3 inhibitors (GSK-3 Inhibitor IX and 1-azakenpaullone), or common environmental contaminants (dibutyl phthalate or the polycyclic aromatic hydrocarbons phenanthrene and fluorene) from the 2 to 8-cell stage through the mid-blastula transition (MBT). These embryos displayed morphological abnormalities at 12.5 h post-fertilization (hpf) that were comparable to embryos exposed to lithium chloride (LiCl) (300 mM LiCl for 10 min, prior to the MBT), a classic disruptor of embryonic axis determination. Whole-mount immunolabeling and laser scanning confocal microscopy were used to localize β-catenin. The commercial GSK-3 Inhibitors as well as LiCl, dibutyl phthalate, fluorene and phenanthrene all induced an increase in the levels of nuclear β-catenin throughout the embryo, indicating that the morphological abnormalities were a result of disruption of Wnt/β-catenin signaling during dorsal-ventral axis specification. The ability of environmental chemicals to directly or indirectly target GSK-3β was assessed. Using Western blot analysis, the ability of these chemicals to affect enzymatic inhibitory phosphorylation at serine 9 on GSK-3β was examined, but no change in the serine phosphorylation state of GSK-3β was detected in exposed embryos. Furthermore, polycyclic aromatic hydrocarbons and dibutyl phthalate had no direct effect on the in vitro kinase activity of GSK-3β. While developmental abnormalities resulting from these axis-disrupting contaminants were linked to β-catenin accumulation in nuclei, the details of the disruption of this signaling pathway remain unknown. Since phenanthrene and fluorene as well as other hydrocarbons have been shown to disrupt axial development in sea urchin embryos, and since axis determination and the Wnt/β-catenin signaling pathway are highly conserved, we propose that these environmental contaminants may impact embryo development through a similar mechanism across phyla.
Collapse
Affiliation(s)
- Elise A Fairbairn
- University of California Davis, Bodega Marine Laboratory, Bodega Bay, CA 94923, USA.
| | | | | |
Collapse
|
19
|
Chen L, Ren F, Zhang H, Wen T, Piao Z, Zhou L, Zheng S, Zhang J, Chen Y, Han Y, Duan Z, Ma Y. Inhibition of glycogen synthase kinase 3β ameliorates D-GalN/LPS-induced liver injury by reducing endoplasmic reticulum stress-triggered apoptosis. PLoS One 2012; 7:e45202. [PMID: 23028846 PMCID: PMC3461002 DOI: 10.1371/journal.pone.0045202] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 08/17/2012] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Glycogen synthase kinase 3β(GSK3β) is a ubiquitous serine-threonine protein kinase that participates in numerous cellular processes and disease pathophysiology. We aimed to determine therapeutic potential of GSK3β inhibition and its mechanism in a well-characterized model of lipopolysaccharide (LPS)-induced model of acute liver failure (ALF). METHODOLOGY In a murine ALF model induced by D-GalN(700 mg/kg)/LPS(10 µg/kg), we analyzed GSK3β mechanisms using a specific chemical inhibitor, SB216763, and detected the role of endoplasmic reticulum stress (ERS). Mice were administered SB216763 at 2 h before or after D-GalN/LPS injection, respectively, and then sacrificed 6 h after D-GalN/LPS treatment to evaluate its prophylactic and therapeutic function. The lethality rate, liver damage, ERS, cytokine expression, MAP kinase, hepatocyte apoptosis and expression of TLR 4 were evaluated, respectively. Whether the inhibition of GSK3β activation protected hepatocyte from ERS-induced apoptosis was investigated in vitro. PRINCIPAL FINDINGS GSK3β became quickly activated (dephosphorylated) upon D-GalN/LPS exposure. Administration of SB216763 not only ameliorated liver injury, as evidenced by reduced transaminase levels, and well-preserved liver architecture, but also decreased lethality. Moreover, GSK3β inhibition resulted in down-regulation of pro-apoptotic proteins C/EBP-homologous protein(CHOP) and caspase-12, which are related to ERS. To further demonstrate the role of ERS, we found that GSK3β inhibition protected hepatocyte from ERS-induced cell death. GSK3β inhibition down-regulated the MAPK pathways, reduced expression of inflammatory cytokines and decreased expression of TLR4. CONCLUSIONS Our findings demonstrate the key function of GSK3β signaling in the pathophysiology of ALF, especially in regulating the ERS, and provide a rationale for targeting GSK3β as a potential therapeutic strategy to ameliorate ALF.
Collapse
Affiliation(s)
- Liyan Chen
- The 2nd Department of Infectious Diseases, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Feng Ren
- Beijing Institute of Liver Diseases, Capital Medical University, Beijing, People’s Republic of China
- Beijing Artificial Liver Treatment & Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Haiyan Zhang
- Beijing Institute of Liver Diseases, Capital Medical University, Beijing, People’s Republic of China
| | - Tao Wen
- Beijing Institute of Liver Diseases, Capital Medical University, Beijing, People’s Republic of China
| | - Zhengfu Piao
- Beijing Institute of Liver Diseases, Capital Medical University, Beijing, People’s Republic of China
| | - Li Zhou
- Beijing Artificial Liver Treatment & Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Sujun Zheng
- Beijing Artificial Liver Treatment & Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jing Zhang
- Beijing Artificial Liver Treatment & Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yu Chen
- Beijing Artificial Liver Treatment & Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yuanping Han
- Beijing Artificial Liver Treatment & Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Zhongping Duan
- Beijing Artificial Liver Treatment & Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yingji Ma
- The Department of Infectious Diseases, The 4th Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| |
Collapse
|
20
|
L. Curry J, A. Torres-Cabala C, L. Warneke C, Zhang P, G. Prieto V. Distinct Subcellular Localization of GSK-3<i>β</i> in Melanocytic Nevi: Implications in Melanocyte Senescence. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/ojpathology.2012.24021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
21
|
Cheong SL, Federico S, Venkatesan G, Mandel AL, Shao YM, Moro S, Spalluto G, Pastorin G. The A3 adenosine receptor as multifaceted therapeutic target: pharmacology, medicinal chemistry, and in silico approaches. Med Res Rev 2011; 33:235-335. [PMID: 22095687 DOI: 10.1002/med.20254] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adenosine is an ubiquitous local modulator that regulates various physiological and pathological functions by stimulating four membrane receptors, namely A(1), A(2A), A(2B), and A(3). Among these G protein-coupled receptors, the A(3) subtype is found mainly in the lung, liver, heart, eyes, and brain in our body. It has been associated with cerebroprotection and cardioprotection, as well as modulation of cellular growth upon its selective activation. On the other hand, its inhibition by selective antagonists has been reported to be potentially useful in the treatment of pathological conditions including glaucoma, inflammatory diseases, and cancer. In this review, we focused on the pharmacology and the therapeutic implications of the human (h)A(3) adenosine receptor (AR), together with an overview on the progress of hA(3) AR agonists, antagonists, allosteric modulators, and radioligands, as well as on the recent advances pertaining to the computational approaches (e.g., quantitative structure-activity relationships, homology modeling, molecular docking, and molecular dynamics simulations) applied to the modeling of hA(3) AR and drug design.
Collapse
Affiliation(s)
- Siew Lee Cheong
- Department of Pharmacy, National University of Singapore, 3 Science Drive 2, Singapore 117543, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Patade VY, Rai AN, Suprasanna P. Expression analysis of sugarcane shaggy-like kinase (SuSK) gene identified through cDNA subtractive hybridization in sugarcane (Saccharum officinarum L.). PROTOPLASMA 2011; 248:613-21. [PMID: 20853012 DOI: 10.1007/s00709-010-0207-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 08/27/2010] [Indexed: 05/20/2023]
Abstract
Identification of genes whose expression enables plants to adapt to any kind of stresses is integral to developing stress tolerance in crop plants. In this study, PCR-based cDNA suppression subtractive hybridization technique was used to construct sugarcane salt (NaCl) stress specific forward and reverse subtracted cDNA library. For this, mRNAs were pooled from the shoot and root tissues stressed with NaCl (200 mM) for various time intervals (0.5 to 18 h). Sequencing the clones from the forward subtracted cDNA library, we identified shaggy-like protein kinase (hereafter referred as sugarcane shaggy-like protein kinase, SuSK; NCBI GenBank EST database Acc: FG804674). The sequence analysis of the SuSK revealed homology to Arabidopsis thaliana shaggy-related protein kinase delta (E value, 1e(-108)), dzeta and iota. Alignment of the catalytic domain sequence of GSK-3/shaggy-like kinase with partial sequence of SuSK performed using ClustalW tool indicated kinase active-site signature sequence. Spatial and temporal transcript expression profiling of the SuSK gene based on Real-Time PCR revealed significant induction of transcript expression in response to short-term salt (NaCl 200 mM) or polyethylene glycol-8,000 (PEG; 20% w/v) induced osmotic stress in leaves and shoots of sugarcane plants. The transcript expression increased progressively under salt stress and reached to 1.5-fold of the control up to 8 h treatment. In response to PEG stress, the transcript expression increased by 1.5-fold over the control in 2-h treatment in leaf, whereas in shoots, the expression remained unchanged in response to the various treatments. Differences in growth parameters, relative water content, and membrane damage rate were statistically insignificant in the short-term salt or PEG-stressed plants as compared to the control, non-stressed plants. Expression analysis revealed the differential and temporal regulation of this gene under salt and PEG stress and that its early induction may indicate involvement in stress signaling.
Collapse
Affiliation(s)
- Vikas Yadav Patade
- Functional Plant Biology Section, Nuclear Agriculture and Biotechnology Division, Bhabha Atomic Research Centre, Trombay, Mumbai, India
| | | | | |
Collapse
|
23
|
Aerts I, Grobben B, Van Ostade X, Slegers H. Cyclic AMP-dependent down regulation of ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (NPP1) in rat C6 glioma. Eur J Pharmacol 2010; 654:1-9. [PMID: 21168404 DOI: 10.1016/j.ejphar.2010.11.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2010] [Revised: 11/11/2010] [Accepted: 11/15/2010] [Indexed: 11/16/2022]
Abstract
In this communication, we demonstrate that an increase in intracellular cAMP by 1) addition of dibutyrylic cAMP (dbcAMP), a membrane-permeable cAMP-analogue, or 2) activation of the β-adrenoceptor with (-)-isoproterenol, down regulates the levels of ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (NPP1) mRNA, NPP1 protein and ecto-NPPase activity in rat C6 glioma cells. DbcAMP and (-)-isoproterenol inhibit NPP1 expression in a time and dose-dependent manner. After 48h of stimulation, 1mM dbcAMP or 5μM (-)-isoproterenol decreases the amount of NPP1 protein by 75±3% and 81±1% respectively. Contrary to down regulation of NPP1, we observe an up regulation of glial fibrillary acidic protein (GFAP), a differentiation marker for astrocytic cells. Using specific inhibitors and activators, we have shown that Ca(2+), PKA, PI 3-K/PKB/GSK-3, Epac/Rap1/PP2A and MAP kinase modules are not involved in the inhibition of NPP1 gene expression. The transcription factor c-jun is significantly reduced while c-fos becomes up regulated after cAMP elevation. However an electrophoretic mobility shift assay with the activator protein-1 motif present in the promoter of the rat NPP1 gene indicates that this motif is not involved in the cAMP-dependent inhibition of NPP1 expression. In conclusion, these results indicate that intracellular cAMP levels regulate the expression of NPP1 in rat C6 glioma cells by a signalling pathway that is different from the GFAP signal transduction pathway.
Collapse
Affiliation(s)
- Indra Aerts
- Department of Biomedical Science, Cellular Biochemistry, Campus Drie Eiken, University of Antwerp, Belgium.
| | | | | | | |
Collapse
|
24
|
Effect of GSK-3 activity, enzymatic inhibition and gene silencing by RNAi on tick oviposition and egg hatching. Parasitology 2010; 137:1537-46. [PMID: 20500916 DOI: 10.1017/s0031182010000284] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is classically described as a key enzyme involved in glycogen metabolism in mammals. It has been shown to be highly conserved among several organisms, mainly in the catalytic domain region. This enzyme has already been described in Rhipicephalus (Boophilus) microplus and the ovaries of females appeared to be the major site of GSK-3 transcription. The treatment with GSK-3 specific inhibitor (alsterpaullone, bromo-indirubin-oxime 6 and indirubin-3-oxime) caused a reduction in oviposition and egg hatching in completely engorged female ticks. The effect was more pronounced in partially engorged females when alsterpaullone was administrated by artificial capillary feeding. Moreover, GSK-3 gene silencing by RNAi in partially engorged females reduced significantly both oviposition and hatching. The study of tick embryogenesis and proteins that participate in this process has been suggested as an important means for the development of novel strategies for parasite control. GSK-3 is an essential protein involved in embryonic processes and for this reason it has already been suggested as a possible antigen candidate for tick control.
Collapse
|
25
|
Molecular distinction between physiological and pathological cardiac hypertrophy: experimental findings and therapeutic strategies. Pharmacol Ther 2010; 128:191-227. [PMID: 20438756 DOI: 10.1016/j.pharmthera.2010.04.005] [Citation(s) in RCA: 642] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiac hypertrophy can be defined as an increase in heart mass. Pathological cardiac hypertrophy (heart growth that occurs in settings of disease, e.g. hypertension) is a key risk factor for heart failure. Pathological hypertrophy is associated with increased interstitial fibrosis, cell death and cardiac dysfunction. In contrast, physiological cardiac hypertrophy (heart growth that occurs in response to chronic exercise training, i.e. the 'athlete's heart') is reversible and is characterized by normal cardiac morphology (i.e. no fibrosis or apoptosis) and normal or enhanced cardiac function. Given that there are clear functional, structural, metabolic and molecular differences between pathological and physiological hypertrophy, a key question in cardiovascular medicine is whether mechanisms responsible for enhancing function of the athlete's heart can be exploited to benefit patients with pathological hypertrophy and heart failure. This review summarizes key experimental findings that have contributed to our understanding of pathological and physiological heart growth. In particular, we focus on signaling pathways that play a causal role in the development of pathological and physiological hypertrophy. We discuss molecular mechanisms associated with features of cardiac hypertrophy, including protein synthesis, sarcomeric organization, fibrosis, cell death and energy metabolism and provide a summary of profiling studies that have examined genes, microRNAs and proteins that are differentially expressed in models of pathological and physiological hypertrophy. How gender and sex hormones affect cardiac hypertrophy is also discussed. Finally, we explore how knowledge of molecular mechanisms underlying pathological and physiological hypertrophy may influence therapeutic strategies for the treatment of cardiovascular disease and heart failure.
Collapse
|
26
|
Wang Z, Havasi A, Gall J, Bonegio R, Li Z, Mao H, Schwartz JH, Borkan SC. GSK3beta promotes apoptosis after renal ischemic injury. J Am Soc Nephrol 2010; 21:284-94. [PMID: 20093356 DOI: 10.1681/asn.2009080828] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The mechanism by which the serine-threonine kinase glycogen synthase kinase-3beta (GSK3beta) affects survival of renal epithelial cells after acute stress is unknown. Using in vitro and in vivo models, we tested the hypothesis that GSK3beta promotes Bax-mediated apoptosis, contributing to tubular injury and organ dysfunction after acute renal ischemia. Exposure of renal epithelial cells to metabolic stress activated GSK3beta, Bax, and caspase 3 and induced apoptosis. Expression of a constitutively active GSK3beta mutant activated Bax and decreased cell survival after metabolic stress. In contrast, pharmacologic inhibition (4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione [TDZD-8]) or RNA interference-mediated knockdown of GSK3beta promoted cell survival. Furthermore, RNA interference-mediated knockdown of Bax abrogated the cell death induced by constitutively active GSK3beta. In a cell-free assay, TDZD-8 inhibited the phosphorylation of a peptide containing the Bax serine(163) site targeted by stress-activated GSK3beta. In rats, TDZD-8 inhibited ischemia-induced activation of GSK3beta, Bax, and caspase 3; ameliorated tubular and epithelial cell damage; and significantly protected renal function. Taken together, GSK3beta-mediated Bax activation induces apoptosis and tubular damage that contribute to acute ischemic kidney injury.
Collapse
Affiliation(s)
- Zhiyong Wang
- Evans Biomedical Research Center, Renal Section, Room 546, 650 Albany Street, Boston, MA 02118-2518, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Hwang ES, Yoon G, Kang HT. A comparative analysis of the cell biology of senescence and aging. Cell Mol Life Sci 2009; 66:2503-24. [PMID: 19421842 PMCID: PMC11115533 DOI: 10.1007/s00018-009-0034-2] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Revised: 04/02/2009] [Accepted: 04/15/2009] [Indexed: 01/10/2023]
Abstract
Various intracellular organelles, such as lysosomes, mitochondria, nuclei, and cytoskeletons, change during replicative senescence, but the utility of these changes as general markers of senescence and their significance with respect to functional alterations have not been comprehensively reviewed. Furthermore, the relevance of these alterations to cellular and functional changes in aging animals is poorly understood. In this paper, we review the studies that report these senescence-associated changes in various aging cells and their underlying mechanisms. Changes associated with lysosomes and mitochondria are found not only in cells undergoing replicative or induced senescence but also in postmitotic cells isolated from aged organisms. In contrast, other changes occur mainly in cells undergoing in vitro senescence. Comparison of age-related changes and their underlying mechanisms in in vitro senescent cells and aged postmitotic cells would reveal the relevance of replicative senescence to the physiological processes occurring in postmitotic cells as individuals age.
Collapse
Affiliation(s)
- Eun Seong Hwang
- Department of Life Science, University of Seoul, Dongdaemungu, Jeonnongdong 90, Seoul 130-743, Republic of Korea.
| | | | | |
Collapse
|
28
|
Expression and activity of glycogen synthase kinase during vitellogenesis and embryogenesis of Rhipicephalus (Boophilus) microplus. Vet Parasitol 2009; 161:261-9. [PMID: 19285806 DOI: 10.1016/j.vetpar.2009.01.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Revised: 01/12/2009] [Accepted: 01/26/2009] [Indexed: 11/24/2022]
Abstract
Glycogen synthase kinase 3 (GSK-3) is classically described as a key enzyme involved in glycogen metabolism in mammals. GSK-3 belongs to a highly conserved family of serine/threonine protein kinases, whose members are involved in hormonal regulation, nuclear signaling, and cell fate determination in higher eukaryotes. We have cloned and characterized the RmGSK-3 gene from Rhipicephalus (Boophilus) microplus tick embryos. DNA and protein sequence analysis depicted high similarity to the corresponding enzyme, from both vertebrate and invertebrate animals. In addition, the mRNA transcription profile identified during embryogenesis was analyzed. We observed that the RmGSK-3 mRNA rapidly decreases from the 1st to 3rd day of development, and increases from the 3rd to 15th day. After the 15th day of development, we observed a near 50% reduction in RmGSK-3 mRNA transcription in comparison to the 1st day. We detected the GSK-3beta isoform in egg homogenates throughout embryogenesis using Western blot analysis. RmGSK-3 mRNA was present in fat body, midgut and ovary from partially and fully engorged adult female ticks. The highest mRNA level was observed in ovaries from both developmental stages and in first-day eggs. Furthermore, RmGSK-3 activity correlated with glycogen content variation. Finally, kinase activity in egg homogenates was inhibited by the specific inhibitor, SB-216763. These data suggest that RmGSK-3beta may be involved in glycogen metabolism regulation during R. microplus embryogenesis.
Collapse
|
29
|
Fishman P, Bar-Yehuda S, Synowitz M, Powell JD, Klotz KN, Gessi S, Borea PA. Adenosine receptors and cancer. Handb Exp Pharmacol 2009:399-441. [PMID: 19639290 PMCID: PMC3598010 DOI: 10.1007/978-3-540-89615-9_14] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The A(1), A(2A), A(2B) and A(3) G-protein-coupled cell surface adenosine receptors (ARs) are found to be upregulated in various tumor cells. Activation of the receptors by specific ligands, agonists or antagonists, modulates tumor growth via a range of signaling pathways. The A(1)AR was found to play a role in preventing the development of glioblastomas. This antitumor effect of the A(1)AR is mediated via tumor-associated microglial cells. Activation of the A(2A)AR results in inhibition of the immune response to tumors via suppression of T regulatory cell function and inhibition of natural killer cell cytotoxicity and tumor-specific CD4+/CD8+ activity. Therefore, it is suggested that pharmacological inhibition of A(2A)AR activation by specific antagonists may enhance immunotherapeutics in cancer therapy. Activation of the A(2B)AR plays a role in the development of tumors via upregulation of the expression levels of angiogenic factors in microvascular endothelial cells. In contrast, it was evident that activation of A(2B)AR results in inhibition of ERK1/2 phosphorylation and MAP kinase activity, which are involved in tumor cell growth signals. Finally, A(3)AR was found to be highly expressed in tumor cells and tissues while low expression levels were noted in normal cells or adjacent tissue. Receptor expression in the tumor tissues was directly correlated to disease severity. The high receptor expression in the tumors was attributed to overexpression of NF-kappaB, known to act as an A(3)AR transcription factor. Interestingly, high A(3)AR expression levels were found in peripheral blood mononuclear cells (PBMCs) derived from tumor-bearing animals and cancer patients, reflecting receptor status in the tumors. A(3)AR agonists were found to induce tumor growth inhibition, both in vitro and in vivo, via modulation of the Wnt and the NF-kappaB signaling pathways. Taken together, A(3)ARs that are abundantly expressed in tumor cells may be targeted by specific A(3)AR agonists, leading to tumor growth inhibition. The unique characteristics of these A(3)AR agonists make them attractive as drug candidates.
Collapse
Affiliation(s)
- P Fishman
- Can-Fite BioPharma, Kiryat Matalon, Petach Tikva, 49170, Israel.
| | | | | | | | | | | | | |
Collapse
|
30
|
De Ketelaere A, Vermeulen L, Vialard J, Van De Weyer I, Van Wauwe J, Haegeman G, Moelans I. Involvement of GSK-3β in TWEAK-mediated NF-κB activation. FEBS Lett 2008; 566:60-4. [PMID: 15147869 DOI: 10.1016/j.febslet.2004.04.041] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2004] [Revised: 03/29/2004] [Accepted: 04/02/2004] [Indexed: 10/26/2022]
Abstract
Glycogen synthase kinase-3beta (GSK-3beta) is a key component of several signaling pathways. We found that a short variant of 'TNF-like weak inducer of apoptosis' (shortTWEAK) formed a complex with GSK-3beta in a yeast two-hybrid system. We demonstrate that shortTWEAK and GSK-3beta colocalize in the nucleus of human neuroblastoma cells. We also show that TWEAK is internalized in different cell lines and that it translocates to the nucleus. This event causes the degradation of IkappaBalpha, the nuclear translocation of both GSK-3beta and p65, and the induction of NF-kappaB-driven gene expression. We demonstrate that the induction of IL-8 expression by TWEAK can be counteracted by LiCl. Taken together, these data suggest that GSK-3beta plays an important role in the signal transduction pathway between TWEAK and NF-kappaB.
Collapse
Affiliation(s)
- Adelheid De Ketelaere
- Enabling Technologies, Johnson & Johnson Pharmaceutical Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | | | | | | | | | | | | |
Collapse
|
31
|
Ni MH, Wu CC, Chan WH, Chien KY, Yu JS. GSK-3 mediates the okadaic acid-induced modification of collapsin response mediator protein-2 in human SK-N-SH neuroblastoma cells. J Cell Biochem 2008; 103:1833-48. [PMID: 17902168 DOI: 10.1002/jcb.21575] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Collapsin response mediator protein-2 (CRMP-2), a phosphoprotein involved in axonal outgrowth and microtubule dynamics, is aberrantly phosphorylated in Alzheimer's disease (AD) brain. Alteration of glycogen synthase kinase-3 (GSK-3) activity is associated with the pathogenesis of AD. Here, we show that CRMP-2 is one of the major substrates for GSK-3 in pig brain extracts. Both GSK-3alpha and 3beta phosphorylate purified pig brain CRMP-2 and significantly alter its mobility in SDS-gels, resembling the CRMP-2 modification observed in AD brain. Interestingly, this modification can be detected in SK-N-SH neuroblastoma cells treated with a phosphatase inhibitor, okadaic acid (OA), and GSK-3 inhibitors completely block this OA-induced event. Knockdown of both GSK-3alpha and 3beta, but not either kinase alone, impairs OA-induced modification of CRMP-2. Mutation of Ser-518 or Ser-522 of CRMP-2, which are highly phosphorylated in AD brain, to Ala blocks the OA-induced modification of CRMP-2 in SK-N-SH cells. Ser-522 prephosphorylated by Cdk5 is required for subsequent GSK-3alpha-mediated phosphorylation of CRMP-2 in vitro. Collectively, our results demonstrate for the first time that OA can induce phosphorylation of CRMP-2 in SK-N-SH cells at sites aberrantly phosphorylated in AD brain, and both GSK-3alpha and 3beta and Ser-522 kinase(s) are involved in this process.
Collapse
Affiliation(s)
- Mei-Hui Ni
- Graduate Institute of Basic Medical Sciences, Medical College of Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| | | | | | | | | |
Collapse
|
32
|
Corena-McLeod MDP, Oliveros A, Charlesworth C, Madden B, Liang YQ, Boules M, Shaw A, Williams K, Richelson E. Paliperidone as a mood stabilizer: a pre-frontal cortex synaptoneurosomal proteomics comparison with lithium and valproic acid after chronic treatment reveals similarities in protein expression. Brain Res 2008; 1233:8-19. [PMID: 18657526 DOI: 10.1016/j.brainres.2008.07.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Revised: 06/20/2008] [Accepted: 07/01/2008] [Indexed: 10/21/2022]
Abstract
A series of recent studies has demonstrated that the molecules involved in regulation of neuronal plasticity are also involved in the mode of action of antidepressants and mood stabilizer drugs. Intracellular calcium signaling, energy metabolism, and neuronal plasticity can be influenced by inducing axonal remodeling and increasing levels of certain synaptic proteins. Because antipsychotic drugs are used as mood stabilizers our studies focused on a newly-marketed antipsychotic drug, paliperidone. We determined changes in rat synaptoneurosomal proteins after chronic treatment with paliperidone, lithium salt, or valproic acid in order to find similarities or differences between the mode of action of paliperidone and these two classical mood stabilizers. We determined differential protein expression profiles in prefrontal cortex (PFC) of male Sprague-Dawley rats (n = 4/group). Synaptoneurosomal-enriched preparations were obtained from PFC after chronic treatment with these three drugs. Proteins were separated by 2D-DIGE and identified by nano-LC-MS/MS. We observed similar protein expression profiles at the synaptoneurosomal level, suggesting that the mode of action for paliperidone is similar to that of lithium and valproic acid. However, the expression profile for paliperidone was more similar to that of lithium. Pathways affected in common by these two drugs included oxidative phosphorylation, electron transport, carbohydrate metabolism, and post-synaptic cytokinesis implicating the effects of these drugs in signaling pathways, energy metabolism, and synaptic plasticity.
Collapse
|
33
|
Simón D, Benitez MJ, Gimenez-Cassina A, Garrido JJ, Bhat RV, Díaz-Nido J, Wandosell F. Pharmacological inhibition of GSK-3 is not strictly correlated with a decrease in tyrosine phosphorylation of residues 216/279. J Neurosci Res 2008; 86:668-74. [PMID: 17893926 DOI: 10.1002/jnr.21523] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Recent evidence suggests that intramolecular autophosphorylation is responsible for the tyrosine phosphorylation (pY) of residues 279 or 216 of glycogen synthase kinase-3 (GSK-3alpha or beta), an event that appears to play an important role in regulating this kinase. This provocative hypothesis was based on the capacity of certain nonselective GSK-3 inhibitors to alter both the activity of GSK-3 and its pY. Inhibitors of GSK-3 are not always capable of preventing this tyrosine phosphorylation, which may require an extended period of time. For example, although lithium chloride inhibits GSK-3 activity, this inhibition does not alter its pY content. Furthermore, even when GSK-3 activity is impaired, GSK-3 pY can still be modified by physiological or pharmacological agents. Taken together, these data indicate that GSK-3 kinase activity is not necessarily correlated with the extent of GSK-3 pY. We hypothesized that some as-yet-unidentified tyrosine kinases and phosphatases may also regulate this kinase.
Collapse
Affiliation(s)
- D Simón
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) y Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
34
|
Claisse G, Charrier B, Kreis M. The Arabidopsis thaliana GSK3/Shaggy like kinase AtSK3-2 modulates floral cell expansion. PLANT MOLECULAR BIOLOGY 2007; 64:113-24. [PMID: 17427040 DOI: 10.1007/s11103-007-9138-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Accepted: 01/17/2007] [Indexed: 05/14/2023]
Abstract
The GSK3/Shaggy family of serine/threonine protein kinases is involved in a series of biological processes in animals, plants and yeast [Charrier et al. (2002) Plant Physiol 130:577-590; Jope and Johnson (2004) Trends Biochem Sci 29:95-102; Li and Nam (2002) Science 295:1299-1301; Piao et al. (2001) Plant J 27:305-314]. In Arabidopsis thaliana, out of the 10 members of the GSK3/Shaggy-like gene family (AtSKs), a biological function has been assigned to only 1 member (AtSK2-1) by mutation. In the present work, a study was undertaken to elucidate the function of AtSK3-2. We have generated mutated versions of the A. thaliana Shaggy-like kinase 3-2 (AtSK3-2), in which Lys(167) and Arg(178), respectively homologues to Lys(85) and Arg(96) of the mammal GSK3beta, were modified into Ala by site-directed mutagenesis. In vitro kinase activity assays of the mutated recombinant protein AtSK3-2-R178A showed that the "primed activity" of the mutated kinase was reduced by 90% while the "non-primed" activity was only 20% reduced compared to the wild-type protein kinase. However, the mutant protein AtSK3-2-K167A showed no activity. Arabidopsis transgenic lines over-expressing AtSK3-2-R178A displayed smaller floral organs, namely pedicels, sepals and petals. Conversely, over-expression of both the wild-type AtSK3-2 protein and the AtSK3-2-K167A mutated version, displayed no altered morphogenesis. Scanning electron microscopic analyses of the AtSK3-2-R178A transgenic plants clearly showed a reduced cell size in flower organs, in which quantitative RT-PCR expression analyses of cell wall expansion enzymes showed reduced transcript levels of three xyloglucan endotransglycosylases (XET), namely XTH22 (TCH4), XTH23 (XTR6) and XTH30 (XTR4). Our data show that AtSK3-2 plays an important role in the control of cell elongation in flower development.
Collapse
Affiliation(s)
- Gaëlle Claisse
- Institut de Biotechnologie des Plantes (IBP), UMR CNRS 8618, Laboratoire de Biologie du Développement des Plantes, Université Paris-Sud XI, Bat 630, Orsay Cedex 91405, France
| | | | | |
Collapse
|
35
|
Giner D, López I, Neco P, Rossetto O, Montecucco C, Gutiérrez LM. Glycogen synthase kinase 3 activation is essential for the snake phospholipase A2 neurotoxin-induced secretion in chromaffin cells. Eur J Neurosci 2007; 25:2341-8. [PMID: 17445232 DOI: 10.1111/j.1460-9568.2007.05497.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Neuroendocrine chromaffin cells were used to study the mechanism of the snake phospholipase A2 (PLA2) neurotoxin enhancement of exocytosis. Notexin, beta-bungarotoxin, taipoxin or textilotoxin enhanced the fast release of catecholamines elicited by flash photolysis of cytosolic caged calcium. Such an increase correlates with the capacity of these neurotoxins to cause fragmentation of the F-actin cortical barrier with subsequent accumulation of vesicles in the proximity of the plasma membrane. These PLA2 neurotoxins do not act via protein kinase C activation, which is known to promote F-actin fragmentation. Lithium, RO31-8220 and SB216763, three inhibitors of the glycogen synthase kinase 3, prevent both the alteration of the F-actin peripheral cortex and the enhancement of fast release elicited by these neurotoxins. In addition, glycogen synthase kinase 3 has been detected by immunolocalization in a membranous compartment of the chromaffin cell endoplasmic reticulum (ER). These results suggest that the activation of this enzyme plays a major role in the enhancement of exocytosis of the readily releasable granules caused by PLA2 neurotoxins in neuroendocrine chromaffin cells.
Collapse
Affiliation(s)
- Daniel Giner
- Instituto de Neurociencias, Centro Mixto CSIC-Universidad Miguel Hernández, Campus de San Juan, 03550 Alicante, Spain
| | | | | | | | | | | |
Collapse
|
36
|
Zhang N, Jiang Y, Zou J, Zhuang S, Jin H, Yu Q. Insights into unbinding mechanisms upon two mutations investigated by molecular dynamics study of GSK3β-axin complex: Role of packing hydrophobic residues. Proteins 2007; 67:941-9. [PMID: 17380482 DOI: 10.1002/prot.21359] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Glycogen synthase kinase 3beta (GSK 3beta) is a key component of several cellular processes including Wnt and insulin signalling pathways. The interaction of GSK3beta with scaffolding peptide axin is thought to be responsible for the effective phosphorylation of beta-catenin, the core effector of Wnt signaling, which has been linked with the occurrence of colon cancer and melanoma. It has been demonstrated that the binding of axin to GSK3beta is abolished by the single-point mutation of Val267 to Gly (V267G) in GSK3beta or Leu392 to Pro (L392P) in axin. Molecular dynamics (MD) simulations were performed on wild type (WT), V267G mutant and L392P one to elucidate the two unbinding mechanisms that occur through different pathways. Besides, rough energy and residue-based energy decomposition were calculated by MM_GBSA (molecular mechanical Generalized_Born surface area) approach to illuminate the instability of the two mutants. The MD simulations of the two mutants and WT reveal that the structure of GSK3beta remains unchanged, while axin moves away from the interfacial hydrophobic pockets in both two mutants. Axin exhibits positional shift in V267G mutant, whereas, losing the hydrogen bonds that are indispensable for stabilizing the helix structure of wild type axin, the helix of axin is distorted in L392P mutant. To conclude, both two mutants destroy the hydrophobic interaction that is essential to the stability of GSK3beta-axin complex.
Collapse
Affiliation(s)
- Na Zhang
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | | | | | | | | | | |
Collapse
|
37
|
Krishnamurthy K, Wang G, Silva J, Condie BG, Bieberich E. Ceramide regulates atypical PKCzeta/lambda-mediated cell polarity in primitive ectoderm cells. A novel function of sphingolipids in morphogenesis. J Biol Chem 2006; 282:3379-90. [PMID: 17105725 DOI: 10.1074/jbc.m607779200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In mammals, the primitive ectoderm is an epithelium of polarized cells that differentiates into all embryonic tissues. Our study shows that in primitive ectoderm cells, the sphingolipid ceramide was elevated and co-distributed with the small GTPase Cdc42 and cortical F-actin at the apicolateral cell membrane. Pharmacological or RNA interference-mediated inhibition of ceramide biosynthesis enhanced apoptosis and impaired primitive ectoderm formation in embryoid bodies differentiated from mouse embryonic stem cells. Primitive ectoderm formation was restored by incubation with ceramide or a ceramide analog. Ceramide depletion prevented plasma membrane translocation of PKCzeta/lambda, its interaction with Cdc42, and phosphorylation of GSK-3beta, a substrate of PKCzeta/lambda. Recombinant PKCzeta formed a complex with the polarity protein Par6 and Cdc42 when bound to ceramide containing lipid vesicles. Our data suggest a novel mechanism by which a ceramide-induced, apicolateral polarity complex with PKCzeta/lambda regulates primitive ectoderm cell polarity and morphogenesis.
Collapse
Affiliation(s)
- Kannan Krishnamurthy
- Institute of Molecular Medicine and Genetics, School of Medicine, Medical College of Georgia, Augusta, Georgia 30912, USA
| | | | | | | | | |
Collapse
|
38
|
García-Alvarez G, Ventura V, Ros O, Aligué R, Gil J, Tauler A. Glycogen synthase kinase-3beta binds to E2F1 and regulates its transcriptional activity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1773:375-82. [PMID: 17050006 DOI: 10.1016/j.bbamcr.2006.09.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Revised: 09/04/2006] [Accepted: 09/05/2006] [Indexed: 01/02/2023]
Abstract
GSK3beta and E2F1 play an important role in the control of proliferation and apoptosis. Previous work has demonstrated that GSK3beta indirectly regulates E2F activity through modulation of cyclin D1 levels. In this work we show that GSK3beta phosphorylates human E2F1 in vitro at serine 403 and threonine 433, both residues localized at its transactivation domain. This phosphorylation was not detected in vivo. However, co-immunoprecipitation experiments do reveal in vivo binding of these proteins. Moreover, uninhibitable and catalitycally inactive GSK3beta forms inhibit the transcriptional activity of a fusion protein containing E2F1 transactivation domain. Both forms of GSK3beta inhibit E2F1 with similar efficiency. Interestingly the effect was independent of the mutation of serine 403 and threonine 433 to alanine. This suggests that this transcriptional modulation is independent of GSK3beta kinase activity and phosphorylation state of serine 403 and threonine 433. The re-targeting of these GSK3beta forms to the nucleus results in a higher capacity to regulate E2F1 transcriptional activity. Depletion of the levels of GSK3beta protein using siRNA activates E2F1 transcriptional activity. The data presented in this study offer a new mechanism of regulation of E2F1 by direct binding of GSK3beta to its transactivation domain.
Collapse
Affiliation(s)
- Gisela García-Alvarez
- Departament de Bioquímica i Biologia Molecular, Facultat de Farmàcia, Universitat de Barcelona. Av. Diagonal 643, E-08028 Barcelona, Catalunya, Spain
| | | | | | | | | | | |
Collapse
|
39
|
Matsuzaki E, Takahashi-Yanaga F, Miwa Y, Hirata M, Watanabe Y, Sato N, Morimoto S, Hirofuji T, Maeda K, Sasaguri T. Differentiation-inducing factor-1 alters canonical Wnt signaling and suppresses alkaline phosphatase expression in osteoblast-like cell lines. J Bone Miner Res 2006; 21:1307-16. [PMID: 16869729 DOI: 10.1359/jbmr.060512] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
UNLABELLED Because DIF-1 has been shown to affect Wnt/beta-catenin signaling pathway, the effects of DIF-1 on osteoblast-like cell lines, SaOS-2 and MC3T3-E1, were examined. We found that DIF-1 inhibited this pathway, resulting in the suppression of ALP promoter activity through the TCF/LEF binding site. INTRODUCTION Differentiation-inducing factor-1 (DIF-1), a morphogen of Dictyostelium, inhibits cell proliferation and induces cell differentiation in several mammalian cells. Previous studies showed that DIF-1 activated glycogen synthase kinase-3beta, suggesting that this chemical could affect the Wnt/beta-catenin signaling pathway. This pathway has been shown to be involved in bone biology. MATERIALS AND METHODS We studied the effects of DIF-1 on SaOS-2 and MC3T3-E1, osteosarcoma cell lines widely used as a model system for ostoblastic cells and murine osteoblast-like cell line, respectively. Reporter gene assays were also carried out to examine the effect of DIF-1 on the Wnt/beta-catenin signaling pathway. RESULTS DIF-1 inhibited SaOS-2 proliferation and reduced alkaline phosphatase (ALP) activity in a concentration- and a time-dependent manner. The expression of ALP was markedly suppressed by DIF-1-treatment in protein and mRNA levels. DIF-1 also suppressed the expression of other osteoblast differentiation markers, including core binding factor alpha1, type I collagen, and osteocalcin, in protein and mRNA levels and inhibited osteoblast-mediated mineralization. Subsequently, we examined the effect of DIF-1 on the Wnt/beta-catenin signaling pathway. We found that DIF-1 suppressed the expression of beta-catenin protein and the activity of the reporter gene containing T-cell factor/lymphoid enhancer-binding factor (TCF/LEF) consensus binding sites. We examined the effect of DIF-1 on a reporter gene driven by the human ALP promoter and found that DIF-1 significantly reduced the ALP reporter gene activity through the TCF/LEF binding site (-1023/-1017 bp). Furthermore, the effect of DIF-1 on MC3T3-E1, a murine osteoblast-like cell line, was examined, and it was found that DIF-1 suppressed ALP mRNA expression by the reduction of the ALP reporter gene activity through the TCF/LEF binding site. CONCLUSIONS Our data suggest that DIF-1 inhibits Wnt/beta-catenin signaling, resulting in the suppression of ALP promoter activity. To our knowledge, this is the first report to analyze the role of the TCF/LEF binding site (-1023/-1017 bp) of the ALP gene promoter in osteoblast-like cell lines.
Collapse
Affiliation(s)
- Etsuko Matsuzaki
- Department of Clinical Pharmacology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Tahimic CGT, Tomimatsu N, Nishigaki R, Fukuhara A, Toda T, Kaibuchi K, Shiota G, Oshimura M, Kurimasa A. Evidence for a role of Collapsin response mediator protein-2 in signaling pathways that regulate the proliferation of non-neuronal cells. Biochem Biophys Res Commun 2006; 340:1244-50. [PMID: 16414354 DOI: 10.1016/j.bbrc.2005.12.132] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2005] [Accepted: 12/18/2005] [Indexed: 11/23/2022]
Abstract
Collapsin response mediator protein-2 or Crmp-2 plays a critical role in the establishment of neuronal polarity. In this study, we present evidence that apart from its functions in neurodevelopment, Crmp-2 is also involved in pathways that regulate the proliferation of non-neuronal cells through its phosphorylation by regulatory proteins. We show that Crmp-2 undergoes dynamic phosphorylation changes in response to contact inhibition-induced quiescence and that hyperphosphorylation of Crmp-2 occurs in a tumor. We further suggest that de-regulation of Crmp-2 phosphorylation levels at certain amino acid residues may lead to aberrant cell proliferation and consequently, tumorigenesis.
Collapse
Affiliation(s)
- Candice Ginn T Tahimic
- Department of Genetic Medicine and Regenerative Therapeutics, Graduate School of Medical Science, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Mori J, Takahashi-Yanaga F, Miwa Y, Watanabe Y, Hirata M, Morimoto S, Shirasuna K, Sasaguri T. Differentiation-inducing factor-1 induces cyclin D1 degradation through the phosphorylation of Thr286 in squamous cell carcinoma. Exp Cell Res 2005; 310:426-33. [PMID: 16153639 DOI: 10.1016/j.yexcr.2005.07.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2004] [Revised: 07/25/2005] [Accepted: 07/29/2005] [Indexed: 11/27/2022]
Abstract
Differentiation-inducing factors (DIFs) are morphogens which induce cell differentiation in Dictyostelium. We reported that DIF-1 and DIF-3 inhibit proliferation and induce differentiation in mammalian cells. In this study, we investigated the effect of DIF-1 on oral squamous cell carcinoma cell lines NA and SAS, well differentiated and poorly differentiated cell lines, respectively. Although DIF-1 did not induce the expression of cell differentiation makers in these cell lines, it inhibited the proliferation of NA and SAS in a dose-dependent manner by restricting the cell cycle in the G0/G1 phase. DIF-1 induced cyclin D1 degradation, but this effect was prevented by treatment with lithium chloride and SB216763, the inhibitors of glycogen synthase kinase-3beta (GSK-3beta). Depletion of endogenous GSK-3beta by RNA interference also attenuated the effect of DIF-1 on cyclin D1 degradation. Therefore, we investigated the effect of DIF-1 on GSK-3beta and found that DIF-1 dephosphorylated GSK-3beta on Ser9 and induced the nuclear translocation of GSK-3beta, suggesting that DIF-1 activated GSK-3beta. Then, we examined the effect of DIF-1 on cyclin D1 mutants (Thr286Ala, Thr288Ala, and Thr286/288Ala). We revealed that Thr286Ala and Thr286/288Ala mutants were highly resistant to DIF-1-induced degradation compared with wild-type cyclin D1, indicating that the phosphorylation of Thr286 was critical for cyclin D1 degradation induced by DIF-1. These results suggest that DIF-1 induces degradation of cyclin D1 through the GSK-3beta-mediated phosphorylation of Thr286.
Collapse
Affiliation(s)
- Jun Mori
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Yasmin T, Takahashi-Yanaga F, Mori J, Miwa Y, Hirata M, Watanabe Y, Morimoto S, Sasaguri T. Differentiation-inducing factor-1 suppresses gene expression of cyclin D1 in tumor cells. Biochem Biophys Res Commun 2005; 338:903-9. [PMID: 16243295 DOI: 10.1016/j.bbrc.2005.10.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2005] [Accepted: 10/06/2005] [Indexed: 11/18/2022]
Abstract
To determine the mechanism by which differentiation-inducing factor-1 (DIF-1), a morphogen of Dictyostelium discoideum, inhibits tumor cell proliferation, we examined the effect of DIF-1 on the gene expression of cyclin D1. DIF-1 strongly reduced the expression of cyclin D1 mRNA and correspondingly decreased the amount of beta-catenin in HeLa cells and squamous cell carcinoma cells. DIF-1 activated glycogen synthase kinase-3beta (GSK-3beta) and inhibition of GSK-3beta attenuated the DIF-1-induced beta-catenin degradation, indicating the involvement of GSK-3beta in this effect. Moreover, DIF-1 reduced the activities of T-cell factor (TCF)/lymphoid enhancer factor (LEF) reporter plasmid and a reporter gene driven by the human cyclin D1 promoter. Eliminating the TCF/LEF consensus site from the cyclin D1 promoter diminished the effect of DIF-1. These results suggest that DIF-1 inhibits Wnt/beta-catenin signaling, resulting in the suppression of cyclin D1 promoter activity.
Collapse
Affiliation(s)
- Tania Yasmin
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Cummings A, Kavlock R. A systems biology approach to developmental toxicology. Reprod Toxicol 2005; 19:281-90. [PMID: 15686864 DOI: 10.1016/j.reprotox.2004.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2004] [Revised: 09/17/2004] [Accepted: 10/09/2004] [Indexed: 11/24/2022]
Abstract
Recent advances in developmental biology have yielded detailed models of gene regulatory networks (GRNs) involved in cell specification and other processes in embryonic differentiation. Such networks form the bedrock on which a systems biology approach to developmental toxicology can be built. In this review, an introduction to GRNs in general is followed by a description of specific networks involved in sea urchin and Drosophila development. A hypothesis is presented regarding the role of GRN analysis in the determination of mechanisms of chemical toxicity during embryonic development. Potential for future directions and research approaches in this area is discussed.
Collapse
Affiliation(s)
- Audrey Cummings
- Reproductive Toxicology Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | | |
Collapse
|
44
|
Sinha D, Wang Z, Ruchalski KL, Levine JS, Krishnan S, Lieberthal W, Schwartz JH, Borkan SC. Lithium activates the Wnt and phosphatidylinositol 3-kinase Akt signaling pathways to promote cell survival in the absence of soluble survival factors. Am J Physiol Renal Physiol 2005; 288:F703-13. [PMID: 15572521 DOI: 10.1152/ajprenal.00189.2004] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mouse proximal tubular cells (BUMPT), when cultured in the absence of growth factors, activate a default apoptotic pathway. Although Wnt signaling antagonizes the effect of proapoptotic triggers, its role in regulating the default pathway of apoptosis is less well defined. The present study examines the hypothesis that lithium (Li+) and (2′Z,3′E)-6-bromoindirubin-3′-oxime (BIO), two glycogen synthase kinase-3β (GSK3β) inhibitors, promote survival of growth factor-deprived renal epithelial cells by activating the Wnt pathway. These studies demonstrate that Li+and BIO activate Wnt signaling as indicated by the following changes: phosphorylation (inhibition) of GSK3β; decreased phosphorylation of β-catenin (a GSK3β substrate); nuclear translocation of β-catenin; specific transcriptional activation of Tcf/catenin-responsive pTopflash constructs; and an increase in the expression of cyclin D1 (indicative of a promitogenic cell response). In addition, Li+or BIO significantly increases the phosphorylation (activation) of Akt, an anti-apoptotic protein, and inhibits apoptosis (decreases both annexin-V staining and caspase-3 activation), during serum deprivation. Inhibition of phosphatidylinositol 3-kinase (responsible for Akt activation) either by wortmanin or LY-294002 prevented Li+- or BIO-induced Akt phosphorylation and reduces cell survival without altering the phosphorylation state of GSK3β. Li+or BIO also increases the expression of insulin-like growth factor-II (IGF-II), a potent proliferative signaling protein. Li+or BIO-free conditioned medium harvested from Li+- or BIO-exposed cells also induced Akt phosphorylation, mimicking the protective effect of the two GSK3β inhibitors on serum-starved cells. Furthermore, the effect of conditioned medium on Akt phosphorylation could be inhibited by either LY-294002 or IGF-binding protein. BIO, a specific GSK3β inhibitor, replicated the protective effect of Li+on cell viability, suggesting that GSK3β activation is important for initiating the apoptotic pathway. Taken together, these data suggest that Li+or BIO promotes renal epithelial cell survival by inhibiting apoptosis through GSK3β-dependent activation of the Wnt pathway and subsequent release of IGF-II. Extracellular IGF-II serves as an autocrine survival factor that is responsible, in part, for activating the anti-apoptotic phosphatidylinositol-3-kinase-Akt pathway during serum deprivation.
Collapse
Affiliation(s)
- Diviya Sinha
- Renal Section, Evans Biomedical Research Center, Rm. 546, 650 Albany St., Boston, MA 02118, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Rentzsch F, Hobmayer B, Holstein TW. Glycogen synthase kinase 3 has a proapoptotic function in Hydra gametogenesis. Dev Biol 2005; 278:1-12. [PMID: 15649456 DOI: 10.1016/j.ydbio.2004.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2003] [Revised: 09/04/2004] [Accepted: 10/05/2004] [Indexed: 11/22/2022]
Abstract
In an approach to study the evolutionary conservation of molecules of the Wnt signal transduction pathway, we analyzed the function of the major negative regulator of this pathway, GSK3 (glycogen synthase kinase 3), in the basal metazoan Hydra. Microinjection assays reveal that HyGSK3 inhibits beta-catenin in zebrafish embryos, indicating that the function of GSK3 in the canonical Wnt signaling pathway is evolutionarily conserved. In Hydra, HyGSK3 transcripts are strongly upregulated during gametogenesis. Treatment of female polyps with the GSK3 inhibitors lithium and alsterpaullone prevents the differentiation of nurse cells and subsequent oocyte formation. Our data indicate that GSK3 is required for the early induction of apoptosis in germline cells, which has been shown to be an initial step in Hydra gametogenesis. Our experiments show that main functions of GSK3 are evolutionarily conserved and unique to multicellular animals, a conclusion which is additionally supported by the presence of specific regulatory domains in the HyGSK3 protein.
Collapse
|
46
|
Litovchick L, Chestukhin A, DeCaprio JA. Glycogen synthase kinase 3 phosphorylates RBL2/p130 during quiescence. Mol Cell Biol 2004; 24:8970-80. [PMID: 15456871 PMCID: PMC517894 DOI: 10.1128/mcb.24.20.8970-8980.2004] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Phosphorylation of the retinoblastoma-related or pocket proteins RB1/pRb, RBL1/p107, and RBL2/p130 regulates cell cycle progression and exit. While all pocket proteins are phosphorylated by cyclin-dependent kinases (CDKs) during the G1/S-phase transition, p130 is also specifically phosphorylated in G0-arrested cells. We have previously identified several phosphorylated residues that match the consensus site for glycogen synthase kinase 3 (GSK3) in the G0 form of p130. Using small-molecule inhibitors of GSK3, site-specific mutants of p130, and phospho-specific antibodies, we demonstrate here that GSK3 phosphorylates p130 during G0. Phosphorylation of p130 by GSK3 contributes to the stability of p130 but does not affect its ability to interact with E2F4 or cyclins. Regulation of p130 by GSK3 provides a novel link between growth factor signaling and regulation of the cell cycle progression and exit.
Collapse
Affiliation(s)
- Larisa Litovchick
- Dana-Farber Cancer Institute, Mayer 457, 44 Binney St., Boston, MA 02115, USA
| | | | | |
Collapse
|
47
|
Jope RS. Inhibition of glycogen synthase kinase-3: a potential therapeutic target of lithium. ACTA ACUST UNITED AC 2004. [DOI: 10.1016/j.cnr.2004.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
48
|
Fishman P, Bar-Yehuda S, Ohana G, Barer F, Ochaion A, Erlanger A, Madi L. An agonist to the A3 adenosine receptor inhibits colon carcinoma growth in mice via modulation of GSK-3 beta and NF-kappa B. Oncogene 2004; 23:2465-71. [PMID: 14691449 DOI: 10.1038/sj.onc.1207355] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A(3) adenosine receptor (A(3)AR) activation with the specific agonist CF101 has been shown to inhibit the development of colon carcinoma growth in syngeneic and xenograft murine models. In the present study, we looked into the effect of CF101 on the molecular mechanisms involved in the inhibition of HCT-116 colon carcinoma in mice. In tumor lesions derived from CF101-treated mice, a decrease in the expression level of protein kinase A (PKA) and an increase in glycogen synthase kinase-3 beta (GSK-3 beta) was observed. This gave rise to downregulation of beta-catenin and its transcriptional gene products cyclin D1 and c-Myc. Further mechanistic studies in vitro revealed that these responses were counteracted by the selective A(3)AR antagonist MRS 1523 and by the GSK-3 beta inhibitors lithium and SB216763, confirming that the observed effects were A(3)AR and GSK-3 beta mediated. CF101 downregulated PKB/Akt expression level, resulting in a decrease in the level and DNA-binding capacity of NF-kappa B, both in vivo and in vitro. Furthermore, the PKA and PKB/Akt inhibitors H89 and Worthmannin mimicked the effect of CF101, supporting their involvement in mediating the response to the agonist. This is the first demonstration that A(3)AR activation induces colon carcinoma growth inhibition via the modulation of the key proteins GSK-3 beta and NF-kappa B.
Collapse
Affiliation(s)
- Pnina Fishman
- Can-Fite Biopharma Ltd, Kiryat-Matalon, Petach-Tikva 49170, Israel.
| | | | | | | | | | | | | |
Collapse
|
49
|
Takahashi-Yanaga F, Shiraishi F, Hirata M, Miwa Y, Morimoto S, Sasaguri T. Glycogen synthase kinase-3beta is tyrosine-phosphorylated by MEK1 in human skin fibroblasts. Biochem Biophys Res Commun 2004; 316:411-5. [PMID: 15020233 DOI: 10.1016/j.bbrc.2004.02.061] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2004] [Indexed: 11/29/2022]
Abstract
Glycogen synthase kinase-3beta (GSK-3beta) can be associated with several proteins in cell. We analyzed the immunoprecipitates by an anti-GSK-3beta antibody from cell lysate of human fibroblasts and found that this protein was co-precipitated with mitogen-activated protein kinase kinase (MEK1/2). U0126, a MEK1/2 inhibitor, inhibited tyrosine phosphorylation of GSK-3beta, suggesting that MEK1/2 was involved in the phosphorylation of Tyr(216) in GSK-3beta. In vitro kinase assay was carried out using a recombinant human active MEK1 and we found that GSK-3beta was phosphorylated on Tyr(216) by this kinase in a dose- and time-dependent manner. Further, the pretreatment of fibroblasts with U0126 inhibited serum-induced nuclear translocation of GSK-3beta. These results suggested that MEK1/2 induces tyrosine phosphorylation of GSK-3beta and this cellular event might induce nuclear translocation of GSK-3beta. This is the first report to suggest that MEK1/2 phosphorylates not only ERK1/2 but also GSK-3beta.
Collapse
Affiliation(s)
- Fumi Takahashi-Yanaga
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
The embryo is initially sexually indifferent, and correct sexual development is dependent on gonadal hormone production. Thus, in the male embryo, anti-Müllerian hormone (AMH), secreted by the Sertoli cells of the testis, induces regression of the Müllerian duct, the anlagen of female reproductive tract. This hormone causes ductal epithelial regression through a paracrine mechanism originating in periductal mesenchyme and the cross-talk between the mesenchymal and epithelial layers accounts for the cranial-to-caudal pattern of Müllerian regression. Here, we review and discuss recent developments concerning the relationship of apoptosis of Müllerian duct to tissue remodeling, mesenchymal-epithelial interactions, and involvement of beta-catenin in AMH signaling in periductal mesenchyme. Determining the role of beta-catenin/LEF-1 signaling is critical for understanding AMH action during Müllerian duct regression.
Collapse
Affiliation(s)
- F Xavier
- Unité de recherches sur l'Endocrinologie du Développement, INSERM, 32 rue des Carnets, 93140 Clamart, France.
| | | |
Collapse
|