1
|
TANG CHAO, ZHONG CHUNYU, ZHU JUNHAO, YUAN FENG, YANG JIN, XU YONG, MA CHIYUAN. GNAS mutations suppress cell invasion by activating MEG3 in growth hormone-secreting pituitary adenoma. Oncol Res 2024; 32:1079-1091. [PMID: 38827318 PMCID: PMC11136687 DOI: 10.32604/or.2024.046007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/08/2023] [Indexed: 06/04/2024] Open
Abstract
Approximately 30%-40% of growth hormone-secreting pituitary adenomas (GHPAs) harbor somatic activating mutations in GNAS (α subunit of stimulatory G protein). Mutations in GNAS are associated with clinical features of smaller and less invasive tumors. However, the role of GNAS mutations in the invasiveness of GHPAs is unclear. GNAS mutations were detected in GHPAs using a standard polymerase chain reaction (PCR) sequencing procedure. The expression of mutation-associated maternally expressed gene 3 (MEG3) was evaluated with RT-qPCR. MEG3 was manipulated in GH3 cells using a lentiviral expression system. Cell invasion ability was measured using a Transwell assay, and epithelial-mesenchymal transition (EMT)-associated proteins were quantified by immunofluorescence and western blotting. Finally, a tumor cell xenograft mouse model was used to verify the effect of MEG3 on tumor growth and invasiveness. The invasiveness of GHPAs was significantly decreased in mice with mutated GNAS compared with that in mice with wild-type GNAS. Consistently, the invasiveness of mutant GNAS-expressing GH3 cells decreased. MEG3 is uniquely expressed at high levels in GHPAs harboring mutated GNAS. Accordingly, MEG3 upregulation inhibited tumor cell invasion, and conversely, MEG3 downregulation increased tumor cell invasion. Mechanistically, GNAS mutations inhibit EMT in GHPAs. MEG3 in mutated GNAS cells prevented cell invasion through the inactivation of the Wnt/β-catenin signaling pathway, which was further validated in vivo. Our data suggest that GNAS mutations may suppress cell invasion in GHPAs by regulating EMT through the activation of the MEG3/Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- CHAO TANG
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - CHUNYU ZHONG
- Department of Neurosurgery, Children’s Hospital of Nanjing Medical University, Nanjing, 210019, China
| | - JUNHAO ZHU
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - FENG YUAN
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - JIN YANG
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - YONG XU
- Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - CHIYUAN MA
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
- Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210029, China
- Department of Neurosurgery, Jinling Hospital, Southern Medical University, Nanjing, 210002, China
- Department of Neurosurgery, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, China
| |
Collapse
|
2
|
Bortolozzi A, Fico G, Berk M, Solmi M, Fornaro M, Quevedo J, Zarate CA, Kessing LV, Vieta E, Carvalho AF. New Advances in the Pharmacology and Toxicology of Lithium: A Neurobiologically Oriented Overview. Pharmacol Rev 2024; 76:323-357. [PMID: 38697859 PMCID: PMC11068842 DOI: 10.1124/pharmrev.120.000007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 05/05/2024] Open
Abstract
Over the last six decades, lithium has been considered the gold standard treatment for the long-term management of bipolar disorder due to its efficacy in preventing both manic and depressive episodes as well as suicidal behaviors. Nevertheless, despite numerous observed effects on various cellular pathways and biologic systems, the precise mechanism through which lithium stabilizes mood remains elusive. Furthermore, there is recent support for the therapeutic potential of lithium in other brain diseases. This review offers a comprehensive examination of contemporary understanding and predominant theories concerning the diverse mechanisms underlying lithium's effects. These findings are based on investigations utilizing cellular and animal models of neurodegenerative and psychiatric disorders. Recent studies have provided additional support for the significance of glycogen synthase kinase-3 (GSK3) inhibition as a crucial mechanism. Furthermore, research has shed more light on the interconnections between GSK3-mediated neuroprotective, antioxidant, and neuroplasticity processes. Moreover, recent advancements in animal and human models have provided valuable insights into how lithium-induced modifications at the homeostatic synaptic plasticity level may play a pivotal role in its clinical effectiveness. We focused on findings from translational studies suggesting that lithium may interface with microRNA expression. Finally, we are exploring the repurposing potential of lithium beyond bipolar disorder. These recent findings on the therapeutic mechanisms of lithium have provided important clues toward developing predictive models of response to lithium treatment and identifying new biologic targets. SIGNIFICANCE STATEMENT: Lithium is the drug of choice for the treatment of bipolar disorder, but its mechanism of action in stabilizing mood remains elusive. This review presents the latest evidence on lithium's various mechanisms of action. Recent evidence has strengthened glycogen synthase kinase-3 (GSK3) inhibition, changes at the level of homeostatic synaptic plasticity, and regulation of microRNA expression as key mechanisms, providing an intriguing perspective that may help bridge the mechanistic gap between molecular functions and its clinical efficacy as a mood stabilizer.
Collapse
Affiliation(s)
- Analia Bortolozzi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Giovanna Fico
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Michael Berk
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Marco Solmi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Michele Fornaro
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Joao Quevedo
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Carlos A Zarate
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Lars V Kessing
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Eduard Vieta
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Andre F Carvalho
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| |
Collapse
|
3
|
Tjeerdema E, Lee Y, Metry R, Hamdoun A. Semi-automated, high-content imaging of drug transporter knockout sea urchin (Lytechinus pictus) embryos. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2024; 342:313-329. [PMID: 38087422 DOI: 10.1002/jez.b.23231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/08/2023] [Accepted: 11/19/2023] [Indexed: 05/01/2024]
Abstract
A defining feature of sea urchins is their extreme fecundity. Urchins produce millions of transparent, synchronously developing embryos, ideal for spatial and temporal analysis of development. This biological feature has been effectively utilized for ensemble measurement of biochemical changes. However, it has been underutilized in imaging studies, where single embryo measurements are used. Here we present an example of how stable genetics and high content imaging, along with machine learning-based image analysis, can be used to exploit the fecundity and synchrony of sea urchins in imaging-based drug screens. Building upon our recently created sea urchin ABCB1 knockout line, we developed a high-throughput assay to probe the role of this drug transporter in embryos. We used high content imaging to compare accumulation and toxicity of canonical substrates and inhibitors of the transporter, including fluorescent molecules and antimitotic cancer drugs, in homozygous knockout and wildtype embryos. To measure responses from the resulting image data, we used a nested convolutional neural network, which rapidly classified embryos according to fluorescence or cell division. This approach identified sea urchin embryos with 99.8% accuracy and determined two-cell and aberrant embryos with 96.3% and 89.1% accuracy, respectively. The results revealed that ABCB1 knockout embryos accumulated the transporter substrate calcein 3.09 times faster than wildtypes. Similarly, knockouts were 4.71 and 3.07 times more sensitive to the mitotic poisons vinblastine and taxol. This study paves the way for large scale pharmacological screens in the sea urchin embryo.
Collapse
Affiliation(s)
- Evan Tjeerdema
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| | - Yoon Lee
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| | - Rachel Metry
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| | - Amro Hamdoun
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
4
|
Yuan Y, Guo M, Gu C, Yang Y. The role of Wnt/β-catenin signaling pathway in the pathogenesis and treatment of multiple myeloma (review). Am J Transl Res 2021; 13:9932-9949. [PMID: 34650674 PMCID: PMC8507016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 06/07/2021] [Indexed: 06/13/2023]
Abstract
Multiple myeloma (MM) is a refractory hematological malignancy characterized by aberrant accumulation of plasma cells. Patients with MM are susceptible to becoming resistant to chemotherapy, eventually leading to relapse. Progression of MM is largely dependent on the bone marrow microenvironment. Stromal cells in the bone marrow microenvironment secrete Wnt ligands to activate Wnt signaling in MM, which is mediated through the transcription regulator β-catenin. In addition, Wnt/β-catenin pathway encourages osteoblast differentiation and bone formation, dysregulation of which is responsible for proliferation and drug resistance of MM cells. As a result, direct inhibition or silencing of β-catenin or associated genes in the Wnt/β-catenin pathway has been proposed to be an effective therapeutic anti-MM strategy. However, the underlying regulatory mechanism of the Wnt/β-catenin pathway in MM remains to be fully elucidated. Herein, we summarized research advances on the specific genes and molecular biology process of Wnt/β-catenin pathway involved in tumorigenesis of MM, as well as the interaction with bone marrow microenvironment. Additionally, comprehensive summaries of drugs or small molecule inhibitors acting on Wnt/β-catenin pathway and targeting MM were introduced. This review intends to provide an overview of theoretical supports for novel Wnt/β-catenin pathway based treatment strategies in MM.
Collapse
Affiliation(s)
- Yuxia Yuan
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese MedicineNanjing 210022, Jiangsu, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
| | - Mengjie Guo
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
| | - Chunyan Gu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese MedicineNanjing 210022, Jiangsu, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
| | - Ye Yang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese MedicineNanjing 210022, Jiangsu, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese MedicineNanjing 210023, Jiangsu, China
| |
Collapse
|
5
|
Deficient LEF1 expression is associated with lithium resistance and hyperexcitability in neurons derived from bipolar disorder patients. Mol Psychiatry 2021; 26:2440-2456. [PMID: 33398088 PMCID: PMC9129103 DOI: 10.1038/s41380-020-00981-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/21/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022]
Abstract
Bipolar disorder (BD) is a psychiatric condition characterized by depressive and manic episodes that affect 2% of the world population. The first-line long-term treatment for mood stabilization is lithium (Li). Induced pluripotent stem cell modeling of BD using hippocampal dentate gyrus-like neurons derived from Li-responsive (LR) and Li-non-responsive (NR) patients previously showed neuronal hyperexcitability. Li treatment reversed hyperexcitability only on the LR neurons. In this study we searched for specific targets of Li resistance in NR neurons and found that the activity of Wnt/β-catenin signaling pathway was severely affected, with a significant decrease in expression of LEF1. Li targets the Wnt/β-catenin signaling pathway by inhibiting GSK-3β and releasing β-catenin that forms a nuclear complex with TCF/LEF1, activating the Wnt/β-catenin transcription program. Therefore, we propose that downregulation of LEF1 may account for Li resistance in NR neurons. Our results show that valproic acid (VPA), a drug used to treat NR patients that also acts downstream of GSK-3β, upregulated LEF1 and Wnt/β-catenin gene targets, increased transcriptional activity of complex β-catenin/TCF/LEF1, and reduced excitability in NR neurons. In addition, decreasing LEF1 expression in control neurons using shLEF1 caused hyperexcitability, confirming that the impact of VPA on excitability in NR neurons was connected to changes in LEF1 and in the Wnt/β-catenin pathway. Our results suggest that LEF1 may be a useful target for the discovery of new drugs for BD treatment.
Collapse
|
6
|
Sinha P, Cree SL, Miller AL, Pearson JF, Kennedy MA. Transcriptional analysis of sodium valproate in a serotonergic cell line reveals gene regulation through both HDAC inhibition-dependent and independent mechanisms. THE PHARMACOGENOMICS JOURNAL 2021; 21:359-375. [PMID: 33649518 DOI: 10.1038/s41397-021-00215-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/17/2021] [Accepted: 01/27/2021] [Indexed: 11/09/2022]
Abstract
Sodium valproate (VPA) is a histone deacetylase (HDAC) inhibitor, widely prescribed in the treatment of bipolar disorder, and yet the precise modes of therapeutic action for this drug are not fully understood. After exposure of the rat serotonergic cell line RN46A to VPA, RNA-sequencing (RNA-Seq) analysis showed widespread changes in gene expression. Analysis by four bioinformatic pipelines revealed as many as 230 genes were significantly upregulated and 72 genes were significantly downregulated. A subset of 23 differentially expressed genes was selected for validation using the nCounter® platform, and of these we obtained robust validation for ADAM23, LSP1, MAOB, MMP13, PAK3, SERPINB2, SNAP91, WNT6, and ZCCHC12. We investigated the effect of lithium on this subset and found four genes, CDKN1C, LSP1, SERPINB2, and WNT6 co-regulated by lithium and VPA. We also explored the effects of other HDAC inhibitors and the VPA analogue valpromide on the subset of 23 selected genes. Expression of eight of these genes, CDKN1C, MAOB, MMP13, NGFR, SHANK3, VGF, WNT6 and ZCCHC12, was modified by HDAC inhibition, whereas others did not appear to respond to several HDAC inhibitors tested. These results suggest VPA may regulate genes through both HDAC-dependent and independent mechanisms. Understanding the broader gene regulatory effects of VPA in this serotonergic cell model should provide insights into how this drug works and whether other HDAC inhibitor compounds may have similar gene regulatory effects, as well as highlighting molecular processes that may underlie regulation of mood.
Collapse
Affiliation(s)
- Priyanka Sinha
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.,Carney Centre for Pharmacogenomics, University of Otago, Christchurch, New Zealand
| | - Simone L Cree
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.,Carney Centre for Pharmacogenomics, University of Otago, Christchurch, New Zealand
| | - Allison L Miller
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.,Carney Centre for Pharmacogenomics, University of Otago, Christchurch, New Zealand
| | - John F Pearson
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.,Carney Centre for Pharmacogenomics, University of Otago, Christchurch, New Zealand.,Biostatistics and Computational Biology Unit, University of Otago, Christchurch, New Zealand
| | - Martin A Kennedy
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand. .,Carney Centre for Pharmacogenomics, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
7
|
Multilevel Regulation of Protein Kinase CδI Alternative Splicing by Lithium Chloride. Mol Cell Biol 2021; 41:e0033820. [PMID: 33288642 PMCID: PMC8088272 DOI: 10.1128/mcb.00338-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Lithium chloride (LiCl) is commonly used in treatment of mood disorders; however, its usage leads to weight gain, which promotes metabolic disorders. Protein kinase C delta (PKCδ), a serine/threonine kinase, is alternatively spliced to PKCδI and PKCδII in 3T3-L1 cells. We previously demonstrated that PKCδI is the predominantly expressed isoform in 3T3-L1 preadipocytes. Here, we demonstrate that LiCl treatment decreases PKCδI levels, increases formation of lipid droplets, and increases oxidative stress. Hence, we investigated the molecular mechanisms underlying the regulation of PKCδI alternative splicing by LiCl. We previously demonstrated that the splice factor SFRS10 is essential for PKCδI splicing. Our results demonstrate that glycogen synthase kinase 3 beta (GSK3β) phosphorylates SFRS10, and SFRS10 is in a complex with long noncoding RNA NEAT1 to promote PKCδI splicing. Using PKCδ splicing minigene and RNA immunoprecipitation assays, our results demonstrate that upon LiCl treatment, NEAT1 levels are reduced, GSK3β activity is inhibited, and SFRS10 phosphorylation is decreased, which leads to decreased expression of PKCδI. Integration of the GSK3β signaling pathway with the ribonucleoprotein complex of long noncoding RNA (lncRNA) NEAT1 and SFRS10 enables fine-tuning of PKCδI expression during adipogenesis. Knowledge of the molecular pathways impacted by LiCl provides an understanding of the ascent of obesity as a comorbidity in disease management.
Collapse
|
8
|
Zhang Q, Zhang Q, Li H, Zhao X, Zhang H. LiCl induces apoptosis via CHOP/NOXA/Mcl-1 axis in human choroidal melanoma cells. Cancer Cell Int 2021; 21:96. [PMID: 33557839 PMCID: PMC7869481 DOI: 10.1186/s12935-021-01778-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/16/2021] [Indexed: 12/22/2022] Open
Abstract
Background Choroidal melanoma is the most common primary intraocular malignancy that occurs in adults. Lithium Chloride Promotes Apoptosis in Human Leukemia NB4 Cells by Inhibiting Glycogen Synthase Kinase-3 Beta. In this study, we aimed to understand whether LiCl exerts anticancer effects on choroidal melanoma cells and elucidate the underlying molecular mechanisms. Methods Human choroidal melanoma cells were treated with LiCl, and cell survival was assessed with MTT assays. Cell reproductive viability was measured by plate colony formation assays. Cell apoptosis was evaluated using flow cytometry, and proteins were detected using western blotting. A human choroidal melanoma xenograft model was established to demonstrate the effect of LiCl on human choroidal melanoma in vivo. Results We found that LiCl inhibited cell survival and clonogenic potential and induced apoptosis in human choroidal melanoma cells. LiCl also reduced the proliferation of choroidal melanoma cells in vivo. Moreover, the upregulation of NOXA and downregulation of Mcl-1 were responsible for LiCl-induced apoptosis. Mcl-1 overexpression obviously impaired LiCl-induced apoptosis and cleavage of caspase8, caspase9, caspase3 and PARP. Moreover, the protein expression of endoplasmic reticulum stress markers, including IRE1α, Bip, p-eIF2α, ATF4 and CHOP, were upregulated following treatment with LiCl. When CHOP expression was knocked down and cells were treated with LiCl, the protein level of NOXA was partially increased, and Mcl-1 expression was increased, while the cleavage of caspase8, caspase9, caspase3 and PARP that was induced by the LiCl was reduced compared with the vehicle treated group. Prolonged ER stress results in the activation of the apoptotic pathway. Conclusions In summary, LiCl induced an endoplasmic reticulum stress response while activating intrinsic apoptosis. Furthermore, the CHOP/NOXA/Mcl-1 axis contributed to LiCl-induced apoptosis both in vitro and in vivo. The present study provides important mechanistic insight into potential cancer treatments involving LiCl and enhances the understanding of human choroidal melanoma.
Collapse
Affiliation(s)
- Qiuqiu Zhang
- Department of Ophthalmology, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong Province, 250033, China.,Department of Ophthalmology, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, China, 277100
| | - Qianwei Zhang
- Department of Ophthalmology, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, China, 277100
| | - Huiyuan Li
- Department of Ophthalmology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250013, China
| | - Xiaofei Zhao
- Department of Ophthalmology, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong Province, 250033, China.
| | - Han Zhang
- Department of Ophthalmology, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong Province, 250033, China.
| |
Collapse
|
9
|
Li J, Gao J, Zhou H, Zhou J, Deng Z, Lu Y, Rao J, Ji G, Gu J, Yang X, Xia Y, Wang X. Inhibition of Glycogen Synthase Kinase 3β Increases the Proportion and Suppressive Function of CD19 +CD24 hiCD27 + Breg Cells. Front Immunol 2020; 11:603288. [PMID: 33343576 PMCID: PMC7746849 DOI: 10.3389/fimmu.2020.603288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 11/02/2020] [Indexed: 01/03/2023] Open
Abstract
CD19+CD24hiCD27+ memory Breg cells exhibit decreased abundance in patients with chronic graft-versus-host disease (cGVHD) after liver transplantation and produce less IL-10 than those from patients without cGVHD and healthy donors. Due to the lack of Breg cells and the difficulty in expanding them in vitro, in mouse models and early human clinical trials, the adoptive transfer of Breg cells to autoimmune diseases is greatly restricted. Glycogen synthase kinase 3β (GSK-3β) is a multifunctional serine/threonine (ser/thr) protein kinase that can participate in B cell growth, metabolic activity, and proliferation. Phosphoprotein array analysis showed that p-GSK-3β-s9 was highly expressed in mBreg cells. Furthermore, here, we demonstrated that GSK-3β expression in mBreg cells is lower than that observed in B cells by flow cytometry. We found that the treatment of B cells with the specific GSK-3β inhibitor SB216763 can significantly increase the proportion and immunosuppressive function of mBreg cells in vitro. Nuclear factor of activated T cells (NFAT) is one of a pivotal regulator of gene expression in adaptive immune system. Here, we observed that inhibition of GSK-3β by SB216763 results in enhanced expression of NFATc1 in B cells, which is essential in regulating the ability of B cells to secrete IL-10. By constructing a xGVHD mouse model, we observed that SB216763-treated mBreg cells effectively prevent xenogeneic GVHD. Here we propose a novel strategy using SB216763 to inhibit GSK-3β and then enhance the proportion and immunosuppressive function of mBreg cells by increasing the expression of NFATc1. This approach may be used as a therapy to ameliorate GVHD and inflammatory diseases.
Collapse
Affiliation(s)
- Jinyang Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, National Health Commission, Nanjing, China
| | - Ji Gao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, National Health Commission, Nanjing, China
| | - Haoming Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, National Health Commission, Nanjing, China
| | - Jinren Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, National Health Commission, Nanjing, China
| | - Zhenghua Deng
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, National Health Commission, Nanjing, China
| | - Yunjie Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, National Health Commission, Nanjing, China.,Hepatopancreatobiliary Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jianhua Rao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, National Health Commission, Nanjing, China
| | - Guwei Ji
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, National Health Commission, Nanjing, China
| | - Jian Gu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, National Health Commission, Nanjing, China
| | - Xinxiang Yang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, National Health Commission, Nanjing, China
| | - Yongxiang Xia
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, National Health Commission, Nanjing, China
| | - Xuehao Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, National Health Commission, Nanjing, China
| |
Collapse
|
10
|
Mitochondria under the spotlight: On the implications of mitochondrial dysfunction and its connectivity to neuropsychiatric disorders. Comput Struct Biotechnol J 2020; 18:2535-2546. [PMID: 33033576 PMCID: PMC7522539 DOI: 10.1016/j.csbj.2020.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/30/2022] Open
Abstract
Neuropsychiatric disorders (NPDs) such as bipolar disorder (BD), schizophrenia (SZ) and mood disorder (MD) are hard to manage due to overlapping symptoms and lack of biomarkers. Risk alleles of BD/SZ/MD are emerging, with evidence suggesting mitochondrial (mt) dysfunction as a critical factor for disease onset and progression. Mood stabilizing treatments for these disorders are scarce, revealing the need for biomarker discovery and artificial intelligence approaches to design synthetically accessible novel therapeutics. Here, we show mt involvement in NPDs by associating 245 mt proteins to BD/SZ/MD, with 7 common players in these disease categories. Analysis of over 650 publications suggests that 245 NPD-linked mt proteins are associated with 800 other mt proteins, with mt impairment likely to rewire these interactions. High dosage of mood stabilizers is known to alleviate manic episodes, but which compounds target mt pathways is another gap in the field that we address through mood stabilizer-gene interaction analysis of 37 prescriptions and over-the-counter psychotropic treatments, which we have refined to 15 mood-stabilizing agents. We show 26 of the 245 NPD-linked mt proteins are uniquely or commonly targeted by one or more of these mood stabilizers. Further, induced pluripotent stem cell-derived patient neurons and three-dimensional human brain organoids as reliable BD/SZ/MD models are outlined, along with multiomics methods and machine learning-based decision making tools for biomarker discovery, which remains a bottleneck for precision psychiatry medicine.
Collapse
|
11
|
Kukharsky MS, Skvortsova VI, Bachurin SO, Buchman VL. In a search for efficient treatment for amyotrophic lateral sclerosis: Old drugs for new approaches. Med Res Rev 2020; 41:2804-2822. [DOI: 10.1002/med.21725] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/23/2020] [Accepted: 08/08/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Michail S. Kukharsky
- Faculty of Medical Biology Pirogov Russian National Research Medical University Moscow Russian Federation
- Institute of Physiologically Active Compounds Russian Academy of Sciences Moscow Region Russian Federation
| | - Veronika I. Skvortsova
- Faculty of Medical Biology Pirogov Russian National Research Medical University Moscow Russian Federation
| | - Sergey O. Bachurin
- Institute of Physiologically Active Compounds Russian Academy of Sciences Moscow Region Russian Federation
| | - Vladimir L. Buchman
- Institute of Physiologically Active Compounds Russian Academy of Sciences Moscow Region Russian Federation
- School of Biosciences Cardiff University Cardiff United Kingdom
| |
Collapse
|
12
|
Exploring lithium's transcriptional mechanisms of action in bipolar disorder: a multi-step study. Neuropsychopharmacology 2020; 45:947-955. [PMID: 31652432 PMCID: PMC7162887 DOI: 10.1038/s41386-019-0556-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/01/2019] [Accepted: 10/17/2019] [Indexed: 12/22/2022]
Abstract
Lithium has been the first-line treatment for bipolar disorder (BD) for more than six decades. Although the molecular effects of lithium have been studied extensively and gene expression changes are generally believed to be involved, the specific mechanisms of action that mediate mood regulation are still not known. In this study, a multi-step approach was used to explore the transcriptional changes that may underlie lithium's therapeutic efficacy. First, we identified genes that are associated both with lithium exposure and with BD, and second, we performed differential expression analysis of these genes in brain tissue samples from BD patients (n = 42) and healthy controls (n = 42). To identify genes that are regulated by lithium exposure, we used high-sensitivity RNA-sequencing of corpus callosum (CC) tissue samples from lithium-treated (n = 8) and non-treated (n = 9) rats. We found that lithium exposure significantly affected 1108 genes (FDR < 0.05), 702 up-regulated and 406 down-regulated. These genes were mostly enriched for molecular functions related to signal transduction, including well-established lithium-related pathways such as mTOR and Wnt signaling. To identify genes with differential expression in BD, we performed expression quantitative trait loci (eQTL) analysis on BD-associated genetic variants from the most recent genome-wide association study (GWAS) using three different gene expression databases. We found 307 unique eQTL genes regulated by BD-associated variants, of which 12 were also significantly modulated by lithium treatment in rats. Two of these showed differential expression in the CC of BD cases: RPS23 was significantly down-regulated (p = 0.0036, fc = 0.80), while GRIN2A showed suggestive evidence of down-regulation in BD (p = 0.056, fc = 0.65). Crucially, GRIN2A was also significantly up-regulated by lithium in the rat brains (p = 2.2e-5, fc = 1.6), which suggests that modulation of GRIN2A expression may be a part of the therapeutic effect of the drug. These results indicate that the recent upsurge in research on this central component of the glutamatergic system, as a target of novel therapeutic agents for affective disorders, is warranted and should be intensified.
Collapse
|
13
|
Pantović-Stefanović M, Petronijević N, Dunjić-Kostić B, Velimirović M, Nikolić T, Jurišić V, Lačković M, Damjanović A, Totić-Poznanović S, Jovanović AA, Ivković M. sVCAM-1, sICAM-1, TNF-α and IL-6 levels in bipolar disorder type I: Acute, longitudinal and therapeutic implications. World J Biol Psychiatry 2019; 19:S41-S51. [PMID: 27841086 DOI: 10.1080/15622975.2016.1259498] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES To explore the serum levels of soluble vascular cell adhesion molecule-1 (sVCAM-1), soluble intercellular cell adhesion molecule-1 (sICAM-1), tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in patients with bipolar disorder (BD), with regard to acute episode characteristics, course of the disorder and treatment. METHODS The study group consisted of 83 patients diagnosed with BD type I. The control group consisted of 73 healthy individuals, matched with the study group according to age, gender and body mass index. The serum levels of sVCAM-1, sICAM-1, TNF-α and IL-6 were measured by ELISA. RESULTS Compared with healthy controls, significantly elevated levels of IL-6 and sICAM-1 and significantly lower levels of TNF-α and sVCAM-1 were identified in acute and remission phases of BD. The acute serum levels of sVCAM-1 were associated with the type and severity of acute mood symptoms as well as with course of illness characteristics. TNF-α was associated with duration of untreated disorder and type of treatment. CONCLUSIONS BD is related to both acute and long-term alterations of immune mediators, including adhesion molecules. The potential immunomodulatory role of pharmacotherapeutic treatment is also to be considered in BD.
Collapse
Affiliation(s)
| | - Nataša Petronijević
- b School of Medicine, University of Belgrade , Belgrade , Serbia.,c Institute of Clinical and Medical Biochemistry , Belgrade , Serbia
| | | | - Milica Velimirović
- b School of Medicine, University of Belgrade , Belgrade , Serbia.,c Institute of Clinical and Medical Biochemistry , Belgrade , Serbia
| | - Tatjana Nikolić
- b School of Medicine, University of Belgrade , Belgrade , Serbia.,c Institute of Clinical and Medical Biochemistry , Belgrade , Serbia
| | - Vladimir Jurišić
- d School of Medicine, University of Kragujevac , Kragujevac , Serbia
| | - Maja Lačković
- a Clinic of Psychiatry , Clinical Centre of Serbia , Belgrade , Serbia.,b School of Medicine, University of Belgrade , Belgrade , Serbia
| | - Aleksandar Damjanović
- a Clinic of Psychiatry , Clinical Centre of Serbia , Belgrade , Serbia.,b School of Medicine, University of Belgrade , Belgrade , Serbia
| | - Sanja Totić-Poznanović
- a Clinic of Psychiatry , Clinical Centre of Serbia , Belgrade , Serbia.,b School of Medicine, University of Belgrade , Belgrade , Serbia
| | - Aleksandar A Jovanović
- a Clinic of Psychiatry , Clinical Centre of Serbia , Belgrade , Serbia.,b School of Medicine, University of Belgrade , Belgrade , Serbia
| | - Maja Ivković
- a Clinic of Psychiatry , Clinical Centre of Serbia , Belgrade , Serbia.,b School of Medicine, University of Belgrade , Belgrade , Serbia
| |
Collapse
|
14
|
Balasubramanian D, Pearson JF, Kennedy MA. Gene expression effects of lithium and valproic acid in a serotonergic cell line. Physiol Genomics 2018; 51:43-50. [PMID: 30576260 DOI: 10.1152/physiolgenomics.00069.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Valproic acid (VPA) and lithium are widely used in the treatment of bipolar disorder. However, the underlying mechanism of action of these drugs is not clearly understood. We used RNA-Seq analysis to examine the global profile of gene expression in a rat serotonergic cell line (RN46A) after exposure to these two mood stabilizer drugs. Numerous genes were differentially regulated in response to VPA (log2 fold change ≥ 1.0; i.e., odds ratio of ≥2, at false discovery rate <5%), but only two genes ( Dynlrb2 and Cdyl2) showed significant differential regulation after exposure of the cells to lithium, with the same analysis criteria. Both of these genes were also regulated by VPA. Many of the differentially expressed genes had functions of potential relevance to mood disorders or their treatment, such as several serpin family genes (including neuroserpin), Nts (neurotensin), Maob (monoamine oxidase B), and Ap2b1, which is important for synaptic vesicle function. Pathway analysis revealed significant enrichment of Gene Ontology terms such as extracellular matrix remodeling, cell adhesion, and chemotaxis. This study in a cell line derived from the raphe nucleus has identified a range of genes and pathways that provide novel insights into potential therapeutic actions of the commonly used mood stabilizer drugs.
Collapse
Affiliation(s)
- Diana Balasubramanian
- Carney Centre for Pharmacogenomics, Department of Pathology and Biomedical Science, University of Otago , Christchurch , New Zealand
| | - John F Pearson
- Carney Centre for Pharmacogenomics, Department of Pathology and Biomedical Science, University of Otago , Christchurch , New Zealand.,Biostatistics and Computational Biology Unit, University of Otago , Christchurch , New Zealand
| | - Martin A Kennedy
- Carney Centre for Pharmacogenomics, Department of Pathology and Biomedical Science, University of Otago , Christchurch , New Zealand
| |
Collapse
|
15
|
Yao R, Sun X, Xie Y, Liu L, Han D, Yao Y, Li H, Li Z, Xu K. Lithium chloride inhibits cell survival, overcomes drug resistance, and triggers apoptosis in multiple myeloma via activation of the Wnt/β-catenin pathway. Am J Transl Res 2018; 10:2610-2618. [PMID: 30210697 PMCID: PMC6129537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 07/07/2018] [Indexed: 06/08/2023]
Abstract
Multiple myeloma (MM) is an extremely malignant plasma cell disease, which is still incurable due to its drug resistance. Lithium chloride (LiCl) functions in many pathological processes, including bipolar disorder, acute brain injuries, and chronic neurodegenerative diseases, but its antagonistic role in MM progression has not been reported thus far. In this study, we found that LiCl inhibited MM cell proliferation and induced MM cell cycle G2/M phase arrest in a dose-dependent manner. Moreover, LiCl overcomes bortezomib (BTZ)-mediated resistance in MM cells and induces apoptosis in BTZ-resistant cells. Our data preliminarily indicate that LiCl induces MM cell apoptosis via activating the Wnt/β-catenin signaling pathway. Overall, our results define LiCl as an inducer of MM cell apoptosis and unveil a crosstalk between BTZ and LiCl in facilitating cell apoptosis.
Collapse
Affiliation(s)
- Ruosi Yao
- Blood Diseases Institute, Xuzhou Medical UniversityXuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Xiaoyang Sun
- Blood Diseases Institute, Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Yu Xie
- Blood Diseases Institute, Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Linlin Liu
- College of Medical Imaging, Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Danyang Han
- Blood Diseases Institute, Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Yao Yao
- Blood Diseases Institute, Xuzhou Medical UniversityXuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Hujun Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical UniversityXuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical UniversityXuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| |
Collapse
|
16
|
de Araujo WM, Robbs BK, Bastos LG, de Souza WF, Vidal FCB, Viola JPB, Morgado-Diaz JA. PTEN Overexpression Cooperates With Lithium to Reduce the Malignancy and to Increase Cell Death by Apoptosis via PI3K/Akt Suppression in Colorectal Cancer Cells. J Cell Biochem 2016. [PMID: 26224641 DOI: 10.1002/jcb.25294] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Lithium is a well-established non-competitive inhibitor of glycogen synthase kinase-3β (GSK-3β), a kinase that is involved in several cellular processes related to cancer progression. GSK-3β is regulated upstream by PI3K/Akt, which is negatively modulated by PTEN. The role that lithium plays in cancer is controversial because lithium can activate or inhibit survival signaling pathways depending on the cell type. In this study, we analyzed the mechanisms by which lithium can modulate events related to colorectal cancer (CRC) progression and evaluated the role that survival signaling pathways such as PI3K/Akt and PTEN play in this context. We show that the administration of lithium decreased the proliferative potential of CRC cells in a GSK-3β-independent manner but induced the accumulation of cells in G2/M phase. Furthermore, high doses of lithium increased apoptosis, which was accompanied by decreased proteins levels of Akt and PTEN. Then, cells that were induced to overexpress PTEN were treated with lithium; we observed that low doses of lithium strongly increased apoptosis. Additionally, PTEN overexpression reduced proliferation, but this effect was minor compared with that in cells treated with lithium alone. Furthermore, we demonstrated that PTEN overexpression and lithium treatment separately reduced cell migration, colony formation, and invasion, and these effects were enhanced when lithium treatment and PTEN overexpression were combined. In conclusion, our findings indicate that PTEN overexpression and lithium treatment cooperate to reduce the malignancy of CRC cells and highlight lithium and PTEN as potential candidates for studies to identify new therapeutic approaches for CRC treatment.
Collapse
Affiliation(s)
- Wallace Martins de Araujo
- Grupo de Biologia Estrutural, Divisão de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 5andar, Rio de Janeiro, Brasil
| | - Bruno Kaufmann Robbs
- Departamento de Ciências Básicas, Campus Universitário de Nova Friburgo, Universidade Federal Fluminense, UFF, Nova Friburgo, Rio de Janeiro, Brasil
| | - Lilian G Bastos
- Grupo de Biologia Estrutural, Divisão de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 5andar, Rio de Janeiro, Brasil
| | - Waldemir F de Souza
- Grupo de Biologia Estrutural, Divisão de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 5andar, Rio de Janeiro, Brasil
| | - Flávia C B Vidal
- Banco de Tumores e DNA do Maranhão, Universidade Federal do Maranhão, Rua Coelho Neto, 311, São Luís, MA, Brasil
| | - João P B Viola
- Grupo de Regulação Gênica, Programa de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 5andar, Rio de Janeiro, Brasil
| | - Jose A Morgado-Diaz
- Grupo de Biologia Estrutural, Divisão de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 5andar, Rio de Janeiro, Brasil
| |
Collapse
|
17
|
Malhi GS, Outhred T. Therapeutic Mechanisms of Lithium in Bipolar Disorder: Recent Advances and Current Understanding. CNS Drugs 2016; 30:931-49. [PMID: 27638546 DOI: 10.1007/s40263-016-0380-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lithium is the most effective and well established treatment for bipolar disorder, and it has a broad array of effects within cellular pathways. However, the specific processes through which therapeutic effects occur and are maintained in bipolar disorder remain unclear. This paper provides a timely update to an authoritative review of pertinent findings that was published in CNS Drugs in 2013. A literature search was conducted using the Scopus database, and was limited by year (from 2012). There has been a resurgence of interest in lithium therapy mechanisms, perhaps driven by technical advancements in recent years that permit the examination of cellular mechanisms underpinning the effects of lithium-along with the reuptake of lithium in clinical practice. Recent research has further cemented glycogen synthase kinase 3β (GSK3β) inhibition as a key mechanism, and the inter-associations between GSK3β-mediated neuroprotective, anti-oxidative and neurotransmission mechanisms have been further elucidated. In addition to highly illustrative cellular research, studies examining higher-order biological systems, such as circadian rhythms, as well as employing innovative animal and human models, have increased our understanding of how lithium-induced changes at the cellular level possibly translate to changes at behavioural and clinical levels. Neural circuitry research is yet to identify clear mechanisms of change in bipolar disorder in response to treatment with lithium, but important structural findings have demonstrated links to the modulation of cellular mechanisms, and peripheral marker and pharmacogenetic studies are showing promising findings that will likely inform the exploration for predictors of lithium treatment response. With a deeper understanding of lithium's therapeutic mechanisms-from the cellular to clinical levels of investigation-comes the opportunity to develop predictive models of lithium treatment response and identify novel drug targets, and recent findings have provided important leads towards these goals.
Collapse
Affiliation(s)
- Gin S Malhi
- Academic Department of Psychiatry, Kolling Institute, Northern Sydney Local Health District, St Leonards, NSW, 2065, Australia. .,Sydney Medical School Northern, The University of Sydney, Sydney, NSW, 2006, Australia. .,CADE Clinic Level 3, Main Hospital Building, Royal North Shore Hospital, Northern Sydney Local Health District, St Leonards, NSW, 2065, Australia.
| | - Tim Outhred
- Academic Department of Psychiatry, Kolling Institute, Northern Sydney Local Health District, St Leonards, NSW, 2065, Australia.,Sydney Medical School Northern, The University of Sydney, Sydney, NSW, 2006, Australia.,CADE Clinic Level 3, Main Hospital Building, Royal North Shore Hospital, Northern Sydney Local Health District, St Leonards, NSW, 2065, Australia
| |
Collapse
|
18
|
Plant N. Can a systems approach produce a better understanding of mood disorders? Biochim Biophys Acta Gen Subj 2016; 1861:3335-3344. [PMID: 27565355 DOI: 10.1016/j.bbagen.2016.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 07/29/2016] [Accepted: 08/22/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND One in twenty-five people suffer from a mood disorder. Current treatments are sub-optimal with poor patient response and uncertain modes-of-action. There is thus a need to better understand underlying mechanisms that determine mood, and how these go wrong in affective disorders. Systems biology approaches have yielded important biological discoveries for other complex diseases such as cancer, and their potential in affective disorders will be reviewed. SCOPE OF REVIEW This review will provide a general background to affective disorders, plus an outline of experimental and computational systems biology. The current application of these approaches in understanding affective disorders will be considered, and future recommendations made. MAJOR CONCLUSIONS Experimental systems biology has been applied to the study of affective disorders, especially at the genome and transcriptomic levels. However, data generation has been slowed by a lack of human tissue or suitable animal models. At present, computational systems biology has only be applied to understanding affective disorders on a few occasions. These studies provide sufficient novel biological insight to motivate further use of computational biology in this field. GENERAL SIGNIFICANCE In common with many complex diseases much time and money has been spent on the generation of large-scale experimental datasets. The next step is to use the emerging computational approaches, predominantly developed in the field of oncology, to leverage the most biological insight from these datasets. This will lead to the critical breakthroughs required for more effective diagnosis, stratification and treatment of affective disorders.
Collapse
Affiliation(s)
- Nick Plant
- School of Bioscience and Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7XH, UK.
| |
Collapse
|
19
|
Breen MS, White CH, Shekhtman T, Lin K, Looney D, Woelk CH, Kelsoe JR. Lithium-responsive genes and gene networks in bipolar disorder patient-derived lymphoblastoid cell lines. THE PHARMACOGENOMICS JOURNAL 2016; 16:446-53. [PMID: 27401222 DOI: 10.1038/tpj.2016.50] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 04/21/2016] [Accepted: 05/18/2016] [Indexed: 12/25/2022]
Abstract
Lithium (Li) is the mainstay mood stabilizer for the treatment of bipolar disorder (BD), although its mode of action is not yet fully understood nor is it effective in every patient. We sought to elucidate the mechanism of action of Li and to identify surrogate outcome markers that can be used to better understand its therapeutic effects in BD patients classified as good (responders) and poor responders (nonresponders) to Li treatment. To accomplish these goals, RNA-sequencing gene expression profiles of lymphoblastoid cell lines (LCLs) were compared between BD Li responders and nonresponders with healthy controls before and after treatment. Several Li-responsive gene coexpression networks were discovered indicating widespread effects of Li on diverse cellular signaling systems including apoptosis and defense response pathways, protein processing and response to endoplasmic reticulum stress. Individual gene markers were also identified, differing in response to Li between BD responders and nonresponders, involved in processes of cell cycle and nucleotide excision repair that may explain part of the heterogeneity in clinical response to treatment. Results further indicated a Li gene expression signature similar to that observed with clonidine treatment, an α2-adrenoceptor agonist. These findings provide a detailed mechanism of Li in LCLs and highlight putative surrogate outcome markers that may permit for advanced treatment decisions to be made and for facilitating recovery in BD patients.
Collapse
Affiliation(s)
- M S Breen
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - C H White
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - T Shekhtman
- Veterans Administration, San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - K Lin
- Department of Affective Disorder, Guangzhou Brain Hospital, Guangzhou Medical University, Guangzhou, China.,Laboratory of Cognition and Emotion, Guangzhou Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - D Looney
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.,Veterans Administration, San Diego Healthcare System, San Diego, CA, USA
| | - C H Woelk
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - J R Kelsoe
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.,Veterans Administration, San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
20
|
Lee YS, Kim JK, Ryu SW, Bae SJ, Kwon K, Noh YH, Kim SY. Integrative meta-analysis of multiple gene expression profiles in acquired gemcitabine-resistant cancer cell lines to identify novel therapeutic biomarkers. Asian Pac J Cancer Prev 2016; 16:2793-800. [PMID: 25854364 DOI: 10.7314/apjcp.2015.16.7.2793] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In molecular-targeted cancer therapy, acquired resistance to gemcitabine is a major clinical problem that reduces its effectiveness, resulting in recurrence and metastasis of cancers. In spite of great efforts to reveal the overall mechanism of acquired gemcitabine resistance, no definitive genetic factors have been identified that are absolutely responsible for the resistance process. Therefore, we performed a cross-platform meta-analysis of three publically available microarray datasets for cancer cell lines with acquired gemcitabine resistance, using the R-based RankProd algorithm, and were able to identify a total of 158 differentially expressed genes (DEGs; 76 up- and 82 down-regulated) that are potentially involved in acquired resistance to gemcitabine. Indeed, the top 20 up- and down-regulated DEGs are largely associated with a common process of carcinogenesis in many cells. For the top 50 up- and down-regulated DEGs, we conducted integrated analyses of a gene regulatory network, a gene co-expression network, and a protein-protein interaction network. The identified DEGs were functionally enriched via Gene Ontology hierarchy and Kyoto Encyclopedia of Genes and Genomes pathway analyses. By systemic combinational analysis of the three molecular networks, we could condense the total number of DEGs to final seven genes. Notably, GJA1, LEF1, and CCND2 were contained within the lists of the top 20 up- or down-regulated DEGs. Our study represents a comprehensive overview of the gene expression patterns associated with acquired gemcitabine resistance and theoretical support for further clinical therapeutic studies.
Collapse
Affiliation(s)
- Young Seok Lee
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, Republic of Korea E-mail :
| | | | | | | | | | | | | |
Collapse
|
21
|
Synaptic Wnt/GSK3β Signaling Hub in Autism. Neural Plast 2016; 2016:9603751. [PMID: 26881141 PMCID: PMC4736967 DOI: 10.1155/2016/9603751] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/29/2015] [Accepted: 11/30/2015] [Indexed: 12/29/2022] Open
Abstract
Hundreds of genes have been associated with autism spectrum disorders (ASDs) and the interaction of weak and de novo variants derive from distinct autistic phenotypes thus making up the “spectrum.” The convergence of these variants in networks of genes associated with synaptic function warrants the study of cell signaling pathways involved in the regulation of the synapse. The Wnt/β-catenin signaling pathway plays a central role in the development and regulation of the central nervous system and several genes belonging to the cascade have been genetically associated with ASDs. In the present paper, we review basic information regarding the role of Wnt/β-catenin signaling in excitatory/inhibitory balance (E/I balance) through the regulation of pre- and postsynaptic compartments. Furthermore, we integrate information supporting the role of the glycogen synthase kinase 3β (GSK3β) in the onset/development of ASDs through direct modulation of Wnt/β-catenin signaling. Finally, given GSK3β activity as key modulator of synaptic plasticity, we explore the potential of this kinase as a therapeutic target for ASD.
Collapse
|
22
|
Lee YS, Hwang SG, Kim JK, Park TH, Kim YR, Myeong HS, Choi JD, Kwon K, Jang CS, Ro YT, Noh YH, Kim SY. Identification of novel therapeutic target genes in acquired lapatinib-resistant breast cancer by integrative meta-analysis. Tumour Biol 2015; 37:2285-97. [PMID: 26361955 DOI: 10.1007/s13277-015-4033-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 09/02/2015] [Indexed: 12/11/2022] Open
Abstract
Acquired resistance to lapatinib is a highly problematic clinical barrier that has to be overcome for a successful cancer treatment. Despite efforts to determine the mechanisms underlying acquired lapatinib resistance (ALR), no definitive genetic factors have been reported to be solely responsible for the acquired resistance in breast cancer. Therefore, we performed a cross-platform meta-analysis of three publically available microarray datasets related to breast cancer with ALR, using the R-based RankProd package. From the meta-analysis, we were able to identify a total of 990 differentially expressed genes (DEGs, 406 upregulated, 584 downregulated) that are potentially associated with ALR. Gene ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of the DEGs showed that "response to organic substance" and "p53 signaling pathway" may be largely involved in ALR process. Of these, many of the top 50 upregulated and downregulated DEGs were found in oncogenesis of various tumors and cancers. For the top 50 DEGs, we constructed the gene coexpression and protein-protein interaction networks from a huge database of well-known molecular interactions. By integrative analysis of two systemic networks, we condensed the total number of DEGs to six common genes (LGALS1, PRSS23, PTRF, FHL2, TOB1, and SOCS2). Furthermore, these genes were confirmed in functional module eigens obtained from the weighted gene correlation network analysis of total DEGs in the microarray datasets ("GSE16179" and "GSE52707"). Our integrative meta-analysis could provide a comprehensive perspective into complex mechanisms underlying ALR in breast cancer and a theoretical support for further chemotherapeutic studies.
Collapse
Affiliation(s)
- Young Seok Lee
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Sun Goo Hwang
- Plant Genomics Laboratory, Department of Applied Plant Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Jin Ki Kim
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Tae Hwan Park
- Department of Plastic and Reconstructive Surgery, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Young Rae Kim
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Ho Sung Myeong
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Jong Duck Choi
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Kang Kwon
- School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Cheol Seong Jang
- Plant Genomics Laboratory, Department of Applied Plant Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Young Tae Ro
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Yun Hee Noh
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Sung Young Kim
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 143-701, Republic of Korea.
| |
Collapse
|
23
|
The effect of depression on serum VEGF level in Alzheimer's disease. DISEASE MARKERS 2015; 2015:742612. [PMID: 25838619 PMCID: PMC4369941 DOI: 10.1155/2015/742612] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/22/2015] [Accepted: 02/24/2015] [Indexed: 12/22/2022]
Abstract
Objective. Growing evidence suggests that angiogenesis might represent a new pathogenic mechanism involved in the progression of Alzheimer's disease (AD). Among angiogenic cytokines, vascular endothelial growth factor (VEGF) levels in AD patients have been evaluated, but the results are controversial among studies. We investigated serum levels of VEGF in AD patients with depression, AD patients without depression, and the controls, respectively. The aim of this study is to elucidate the relationship between VEGF, depression, and cognitive impairment in AD. Methods. The CDR (Clinical Dementia Rating), MMSE-KC (the Mini-Mental Status Examination-Korean version), and SGDS-K (the Korean version of the Geriatric Depression Scale-Short Form) were measured in the subjects. Serum VEGF levels were measured in 24 AD patients with depression, 25 AD patients without depression, and 26 controls, using an enzyme-linked immunosorbent assay kit. Results. Serum VEGF levels in AD patients with depression were significantly higher than AD patients without depression or the control. A correlation was observed between VEGF and scores on SGDS-K, but no correlation was detected between VEGF and MMSE-KC scores. Conclusion. Serum VEGF levels in AD patients with depression were higher than those without depression. Depression might be associated with changes in serum levels of VEGF in AD patients.
Collapse
|
24
|
Li L, Song H, Zhong L, Yang R, Yang XQ, Jiang KL, Liu BZ. Lithium Chloride Promotes Apoptosis in Human Leukemia NB4 Cells by Inhibiting Glycogen Synthase Kinase-3 Beta. Int J Med Sci 2015; 12:805-10. [PMID: 26516309 PMCID: PMC4615241 DOI: 10.7150/ijms.12429] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 09/02/2015] [Indexed: 01/10/2023] Open
Abstract
Acute promyelocytic leukemia (APL) is a subtype of acute myeloid leukemia (AML). With the application of all-trans retinoic acid (ATRA) and arsenic trioxide (ATO), APL becomes one of best prognosis of leukemia. However, ATRA and ATO are not effective against all APLs. Therefore, a new strategy for APL treatment is necessary. Here, we investigated whether lithium chloride (LiCl), a drug used for the treatment of mental illness, could promote apoptosis in human leukemia NB4 cells. We observed that treatment with LiCl significantly accelerated apoptosis in NB4 cells and led to cell cycle arrest at G2/M phase. Moreover, LiCl significantly increased the level of Ser9-phosphorylated glycogen synthase kinase 3β(p-GSK-3β), and decreased the level of Akt1 protein in a dose-dependent manner. In addition, LiCl inhibition of c-Myc also enhanced cell death with a concomitant increase in β-catnin. Taken together, these findings demonstrated that LiCl promoted apoptosis in NB4 cells through the Akt signaling pathway and that G2/M phase arrest was induced by increase of p-GSK-3β(S9).
Collapse
Affiliation(s)
- Liu Li
- 1. Central Laboratory of Yong-chuan hospital, Chongqing Medical University, Chongqing 402160, China. ; 2. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chong-qing Medical University, Chongqing 400016, China
| | - Hao Song
- 1. Central Laboratory of Yong-chuan hospital, Chongqing Medical University, Chongqing 402160, China
| | - Liang Zhong
- 2. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chong-qing Medical University, Chongqing 400016, China
| | - Rong Yang
- 2. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chong-qing Medical University, Chongqing 400016, China
| | - Xiao-Qun Yang
- 2. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chong-qing Medical University, Chongqing 400016, China
| | - Kai-Ling Jiang
- 2. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chong-qing Medical University, Chongqing 400016, China
| | - Bei-Zhong Liu
- 1. Central Laboratory of Yong-chuan hospital, Chongqing Medical University, Chongqing 402160, China. ; 2. Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chong-qing Medical University, Chongqing 400016, China
| |
Collapse
|
25
|
Scheuing L, Chiu CT, Liao HM, Linares GR, Chuang DM. Preclinical and clinical investigations of mood stabilizers for Huntington's disease: what have we learned? Int J Biol Sci 2014; 10:1024-38. [PMID: 25285035 PMCID: PMC4183923 DOI: 10.7150/ijbs.9898] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 07/08/2014] [Indexed: 12/20/2022] Open
Abstract
Huntington's disease (HD) is a lethal, autosomal dominant neurodegenerative disorder caused by CAG repeat expansions at exon 1 of the huntingtin (Htt) gene, which encodes for a mutant huntingtin protein (mHtt). Prominent symptoms of HD include motor dysfunction, characterized by chorea; psychiatric disturbances such as mood and personality changes; and cognitive decline that may lead to dementia. Pathologically multiple complex processes and pathways are involved in the development of HD, including selective loss of neurons in the striatum and cortex, dysregulation of cellular autophagy, mitochondrial dysfunction, decreased neurotrophic and growth factor levels, and aberrant regulation of gene expression and epigenetic patterns. No cure for HD presently exists, nor are there drugs that can halt the progression of this devastating disease. Therefore, the need to discover neuroprotective modalities to combat HD is critical. In basic and preclinical studies using cellular and animal HD models, the mood stabilizers lithium and valproic acid (VPA) have shown multiple beneficial effects, including behavioral and motor improvement, enhanced neuroprotection, and lifespan extension. Recent studies in transgenic HD mice support the notion that combined lithium/VPA treatment is more effective than treatment with either drug alone. In humans, several clinical studies of HD patients found that lithium treatment improved mood, and that VPA treatment both stabilized mood and moderately reduced chorea. In contrast, other studies observed that the hallmark features of HD were unaffected by treatment with either lithium or VPA. The current review discusses preclinical and clinical investigations of the beneficial effects of lithium and VPA on HD pathophysiology.
Collapse
Affiliation(s)
- Lisa Scheuing
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Drive MSC 1363, Bethesda, MD 20892-1363, USA
| | - Chi-Tso Chiu
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Drive MSC 1363, Bethesda, MD 20892-1363, USA
| | - Hsiao-Mei Liao
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Drive MSC 1363, Bethesda, MD 20892-1363, USA
| | - Gabriel R Linares
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Drive MSC 1363, Bethesda, MD 20892-1363, USA
| | - De-Maw Chuang
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Drive MSC 1363, Bethesda, MD 20892-1363, USA
| |
Collapse
|
26
|
Thangavel M, Seelan RS, Lakshmanan J, Vadnal RE, Stagner JI, Parthasarathy LK, Casanova MF, El-Mallakh RS, Parthasarathy RN. Proteomic analysis of rat prefrontal cortex after chronic valproate treatment. J Neurosci Res 2014; 92:927-36. [DOI: 10.1002/jnr.23373] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 01/28/2014] [Accepted: 01/28/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Muthusamy Thangavel
- Molecular Neuroscience and Bioinformatics Laboratories; Mental Health; Behavioral Science; and Research Services; Robley Rex Veterans Affairs Medical Center; Louisville Kentucky
- Department of Psychiatry and Behavioral Sciences; University of Louisville; Louisville Kentucky
| | - Ratnam S. Seelan
- Molecular Neuroscience and Bioinformatics Laboratories; Mental Health; Behavioral Science; and Research Services; Robley Rex Veterans Affairs Medical Center; Louisville Kentucky
- Department of Psychiatry and Behavioral Sciences; University of Louisville; Louisville Kentucky
- Department of Molecular; Cellular; and Craniofacial Biology; School of Dentistry, University of Louisville; Louisville Kentucky
| | - Jaganathan Lakshmanan
- Molecular Neuroscience and Bioinformatics Laboratories; Mental Health; Behavioral Science; and Research Services; Robley Rex Veterans Affairs Medical Center; Louisville Kentucky
- Price Institute of Surgical Research; Department of Surgery; School of Medicine, University of Louisville; Louisville Kentucky
| | - Robert E. Vadnal
- Eastern Colorado Health Care System; Department of Veterans Affairs; Pueblo Colorado
| | - John I. Stagner
- Molecular Neuroscience and Bioinformatics Laboratories; Mental Health; Behavioral Science; and Research Services; Robley Rex Veterans Affairs Medical Center; Louisville Kentucky
| | - Latha K. Parthasarathy
- Molecular Neuroscience and Bioinformatics Laboratories; Mental Health; Behavioral Science; and Research Services; Robley Rex Veterans Affairs Medical Center; Louisville Kentucky
- Department of Psychiatry and Behavioral Sciences; University of Louisville; Louisville Kentucky
| | - Manuel F. Casanova
- Department of Psychiatry and Behavioral Sciences; University of Louisville; Louisville Kentucky
| | - Rifaat Shody El-Mallakh
- Department of Psychiatry and Behavioral Sciences; University of Louisville; Louisville Kentucky
| | - Ranga N. Parthasarathy
- Molecular Neuroscience and Bioinformatics Laboratories; Mental Health; Behavioral Science; and Research Services; Robley Rex Veterans Affairs Medical Center; Louisville Kentucky
- Department of Psychiatry and Behavioral Sciences; University of Louisville; Louisville Kentucky
- Department of Biochemistry and Molecular Biology; University of Louisville; Louisville Kentucky
| |
Collapse
|
27
|
Wu R, Fan J, Zhao J, Calabrese JR, Gao K. The relationship between neurotrophins and bipolar disorder. Expert Rev Neurother 2013; 14:51-65. [DOI: 10.1586/14737175.2014.863709] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
28
|
Calati R, Crisafulli C, Balestri M, Serretti A, Spina E, Calabrò M, Sidoti A, Albani D, Massat I, Höfer P, Amital D, Juven-Wetzler A, Kasper S, Zohar J, Souery D, Montgomery S, Mendlewicz J. Evaluation of the role of MAPK1 and CREB1 polymorphisms on treatment resistance, response and remission in mood disorder patients. Prog Neuropsychopharmacol Biol Psychiatry 2013; 44:271-8. [PMID: 23537502 DOI: 10.1016/j.pnpbp.2013.03.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 02/11/2013] [Accepted: 03/17/2013] [Indexed: 01/21/2023]
Abstract
Treatment resistant depression (TRD) is a significant clinical and public health problem. Among others, neuroplasticity and inflammatory pathways seem to play a crucial role in the pathomechanisms of antidepressant efficacy. The primary aim of this study was to investigate whether a set of single nucleotide polymorphisms (SNPs) within two genes implicated in neuroplasticity and inflammatory processes (the mitogen activated protein kinase 1, MAPK1 (rs3810608, rs6928, rs13515 and rs8136867), and the cyclic AMP responsive element binding protein 1, CREB1 (rs889895, rs6740584, rs2551922 and rs2254137)) was associated with antidepressant treatment resistance (according to two different definitions), in 285 Major Depressive Disorder (MDD) patients. As secondary aims, we investigated the genetic modulation of the same SNPs on response, remission and other clinical features both in MDD patients and in a larger sample including 82 Bipolar Disorder (BD) patients as well. All patients were screened in the context of a European multicenter project. No association between both the investigated genes and treatment resistance and response was found in MDD patients. However, considering remission, higher rates of CREB1 rs889895 GG genotype were reported in MDD patients. Moreover, MAPK1 rs8136867 AG genotype was found to be associated with remission in the whole sample (MDD and BD). Present results suggest that some genetic polymorphisms in both CREB1 and MAPK1 could be associated with treatment remission. Although further research is needed to draw more definitive conclusions, such results are intriguing since suggest a potential role of two genes implicated in neuroplasticity and inflammatory processes in symptom remission after antidepressant treatment.
Collapse
Affiliation(s)
- Raffaella Calati
- IRCCS Centro S. Giovanni di Dio, Fatebenefratelli, Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Lan Y, Liu X, Zhang R, Wang K, Wang Y, Hua ZC. Lithium enhances TRAIL-induced apoptosis in human lung carcinoma A549 cells. Biometals 2013; 26:241-54. [PMID: 23378009 DOI: 10.1007/s10534-012-9607-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 12/28/2012] [Indexed: 12/21/2022]
Abstract
Non-small cell lung cancer (NSCLC) A549 cells are resistant to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis. Therefore, combination therapy using sensitizing agents to overcome TRAIL resistance may provide new strategies for treatment of NSCLC. Here, we investigated whether lithium chloride (LiCl), a drug for mental illness, could sensitize A549 cells to TRAIL-induced apoptosis. We observed that LiCl significantly enhanced A549 cells apoptosis through up-regulation of death receptors DR4 and DR5 and activation of caspase cascades. In addition, G2/M arrest induced by LiCl also contributed to TRAIL-induced apoptosis. Concomitantly, LiCl strongly inhibited the activity of c-Jun N-terminal kinases (JNKs), and the inhibition of JNKs by SP600125 also induced G2/M arrest and augmented cell death caused by TRAIL or TRAIL plus LiCl. However, glycogen synthase kinase-3β (GSK3β) inhibition was not involved in TRAIL sensitization induced by LiCl. Collectively, these findings indicated that LiCl sensitized A549 cells to TRAIL-induced apoptosis through caspases-dependent apoptotic pathway via death receptors signaling and G2/M arrest induced by inhibition of JNK activation, but independent of GSK3β.
Collapse
Affiliation(s)
- Yan Lan
- The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210093, China
| | | | | | | | | | | |
Collapse
|
30
|
Chiu CT, Wang Z, Hunsberger JG, Chuang DM. Therapeutic potential of mood stabilizers lithium and valproic acid: beyond bipolar disorder. Pharmacol Rev 2013; 65:105-42. [PMID: 23300133 PMCID: PMC3565922 DOI: 10.1124/pr.111.005512] [Citation(s) in RCA: 282] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The mood stabilizers lithium and valproic acid (VPA) are traditionally used to treat bipolar disorder (BD), a severe mental illness arising from complex interactions between genes and environment that drive deficits in cellular plasticity and resiliency. The therapeutic potential of these drugs in other central nervous system diseases is also gaining support. This article reviews the various mechanisms of action of lithium and VPA gleaned from cellular and animal models of neurologic, neurodegenerative, and neuropsychiatric disorders. Clinical evidence is included when available to provide a comprehensive perspective of the field and to acknowledge some of the limitations of these treatments. First, the review describes how action at these drugs' primary targets--glycogen synthase kinase-3 for lithium and histone deacetylases for VPA--induces the transcription and expression of neurotrophic, angiogenic, and neuroprotective proteins. Cell survival signaling cascades, oxidative stress pathways, and protein quality control mechanisms may further underlie lithium and VPA's beneficial actions. The ability of cotreatment to augment neuroprotection and enhance stem cell homing and migration is also discussed, as are microRNAs as new therapeutic targets. Finally, preclinical findings have shown that the neuroprotective benefits of these agents facilitate anti-inflammation, angiogenesis, neurogenesis, blood-brain barrier integrity, and disease-specific neuroprotection. These mechanisms can be compared with dysregulated disease mechanisms to suggest core cellular and molecular disturbances identifiable by specific risk biomarkers. Future clinical endeavors are warranted to determine the therapeutic potential of lithium and VPA across the spectrum of central nervous system diseases, with particular emphasis on a personalized medicine approach toward treating these disorders.
Collapse
Affiliation(s)
- Chi-Tso Chiu
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | |
Collapse
|
31
|
Graae L, Karlsson R, Paddock S. Significant association of estrogen receptor binding site variation with bipolar disorder in females. PLoS One 2012; 7:e32304. [PMID: 22389694 PMCID: PMC3289647 DOI: 10.1371/journal.pone.0032304] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 01/26/2012] [Indexed: 12/22/2022] Open
Abstract
Major depression is nearly twice as prevalent in women compared to men. In bipolar disorder, depressive episodes have been reported to be more common amongst female patients. Furthermore, periods of depression often correlate with periods of hormonal fluctuations. A link between hormone signaling and these mood disorders has, therefore, been suggested to exist in many studies. Estrogen, one of the primary female sex hormones, mediates its effect mostly by binding to estrogen receptors (ERs). Nuclear ERs function as transcription factors and regulate gene transcription by binding to specific DNA sequences. A nucleotide change in the binding sequence might alter the binding efficiency, which could affect transcription levels of nearby genes. In order to investigate if variation in ER DNA-binding sequences may be involved in mood disorders, we conducted a genome-wide study of ER DNA-binding in patients diagnosed with major depression or bipolar disorder. Association studies were performed within each gender separately and the results were corrected for multiple testing by the Bonferroni method. In the female bipolar disorder material a significant association result was found for rs6023059 (corrected p-value = 0.023; odds ratio (OR) 0.681, 95% confidence interval (CI) 0.570–0.814), a single nucleotide polymorphism (SNP) placed downstream of the gene coding for transglutaminase 2 (TGM2). Thus, females with a specific genotype at this SNP may be more vulnerable to fluctuating estrogen levels, which may then act as a triggering factor for bipolar disorder.
Collapse
Affiliation(s)
- Lisette Graae
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | | | | |
Collapse
|
32
|
Lee BH, Kim YK. Increased plasma VEGF levels in major depressive or manic episodes in patients with mood disorders. J Affect Disord 2012; 136:181-184. [PMID: 21862441 DOI: 10.1016/j.jad.2011.07.021] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2011] [Revised: 07/24/2011] [Accepted: 07/24/2011] [Indexed: 11/16/2022]
Abstract
BACKGROUND The neurotrophic hypothesis suggests that mood disorders are associated with dysfunction of neuronal networks under the influence of neurotrophic factors. Vascular endothelial growth factor (VEGF) is a neurotrophic factor as well as an angiogenic cytokine. METHODS We examined plasma VEGF levels in 35 unipolar patients who were diagnosed with current major depressive disorder (MDD), 35 bipolar patients who were diagnosed with bipolar I disorder, manic episode (BM), and 60 healthy controls. The severity of depressive or manic symptoms was measured using the Hamilton Depression Rating Scale (HDRS) or the Young Mania rating scale (YMRS), respectively. RESULTS Plasma VEGF levels were 163.28±135.33 pg/mL in MDD patients, 199.82±182.59 pg/mL in BM patients, and 110.05±109.57 pg/mL in healthy controls. Both MDD and BM patients had significantly higher VEGF levels than healthy controls when controlling for BMI as a covariate (p=0.010). Patients' VEGF levels were not correlated with either HDRS or YMRS scores. LIMITATIONS We assessed plasma VEGF levels at one time point, and we did not determine the source of VEGF in our samples. CONCLUSIONS Plasma VEGF levels were elevated in patients with acute episodes of major depressive disorder and bipolar disorder. Such an alteration of VEGF in acute episode, mood disorders may be associated with a neuroprotective role for VEGF.
Collapse
Affiliation(s)
- Bun-Hee Lee
- Department of Psychiatry, College of Medicine, Korea University, Seoul, Republic of Korea; KARF Hospital, the Korean Alcohol Research Foundation, Gyeonggido, Republic of Korea
| | - Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University, Seoul, Republic of Korea; Division of Brain Korea 21 Biomedical Science, Korea University, Republic of Korea.
| |
Collapse
|