1
|
Phan BN, Ray MH, Xue X, Fu C, Fenster RJ, Kohut SJ, Bergman J, Haber SN, McCullough KM, Fish MK, Glausier JR, Su Q, Tipton AE, Lewis DA, Freyberg Z, Tseng GC, Russek SJ, Alekseyev Y, Ressler KJ, Seney ML, Pfenning AR, Logan RW. Single nuclei transcriptomics in human and non-human primate striatum in opioid use disorder. Nat Commun 2024; 15:878. [PMID: 38296993 PMCID: PMC10831093 DOI: 10.1038/s41467-024-45165-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/12/2024] [Indexed: 02/02/2024] Open
Abstract
In brain, the striatum is a heterogenous region involved in reward and goal-directed behaviors. Striatal dysfunction is linked to psychiatric disorders, including opioid use disorder (OUD). Striatal subregions are divided based on neuroanatomy, each with unique roles in OUD. In OUD, the dorsal striatum is involved in altered reward processing, formation of habits, and development of negative affect during withdrawal. Using single nuclei RNA-sequencing, we identified both canonical (e.g., dopamine receptor subtype) and less abundant cell populations (e.g., interneurons) in human dorsal striatum. Pathways related to neurodegeneration, interferon response, and DNA damage were significantly enriched in striatal neurons of individuals with OUD. DNA damage markers were also elevated in striatal neurons of opioid-exposed rhesus macaques. Sex-specific molecular differences in glial cell subtypes associated with chronic stress were found in OUD, particularly female individuals. Together, we describe different cell types in human dorsal striatum and identify cell type-specific alterations in OUD.
Collapse
Affiliation(s)
- BaDoi N Phan
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
- Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Madelyn H Ray
- Department of Pharmacology, Physiology & Biophysics, Boston University School of Medicine, Boston, MA, 02118, USA
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Chen Fu
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Robert J Fenster
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Division of Depression and Anxiety, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
| | - Stephen J Kohut
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Behavioral Biology Program, McLean Hospital, Belmont, MA, 02478, USA
| | - Jack Bergman
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Behavioral Biology Program, McLean Hospital, Belmont, MA, 02478, USA
| | - Suzanne N Haber
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine, Rochester, NY, 14642, USA
| | - Kenneth M McCullough
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Madeline K Fish
- Center for Systems Neuroscience, Boston University, Boston, MA, 02118, USA
- Graduate Program for Neuroscience, Boston University, Boston, MA, 02118, USA
| | - Jill R Glausier
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Qiao Su
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Allison E Tipton
- Center for Systems Neuroscience, Boston University, Boston, MA, 02118, USA
- Graduate Program for Neuroscience, Boston University, Boston, MA, 02118, USA
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Shelley J Russek
- Department of Pharmacology, Physiology & Biophysics, Boston University School of Medicine, Boston, MA, 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, 02118, USA
- Graduate Program for Neuroscience, Boston University, Boston, MA, 02118, USA
| | - Yuriy Alekseyev
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Division of Depression and Anxiety, McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA
| | - Marianne L Seney
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Andreas R Pfenning
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
| | - Ryan W Logan
- Department of Pharmacology, Physiology & Biophysics, Boston University School of Medicine, Boston, MA, 02118, USA.
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
2
|
Mackiewicz J, Lisek M, Boczek T. Targeting CaN/NFAT in Alzheimer's brain degeneration. Front Immunol 2023; 14:1281882. [PMID: 38077352 PMCID: PMC10701682 DOI: 10.3389/fimmu.2023.1281882] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive loss of cognitive functions. While the exact causes of this debilitating disorder remain elusive, numerous investigations have characterized its two core pathologies: the presence of β-amyloid plaques and tau tangles. Additionally, multiple studies of postmortem brain tissue, as well as results from AD preclinical models, have consistently demonstrated the presence of a sustained inflammatory response. As the persistent immune response is associated with neurodegeneration, it became clear that it may also exacerbate other AD pathologies, providing a link between the initial deposition of β-amyloid plaques and the later development of neurofibrillary tangles. Initially discovered in T cells, the nuclear factor of activated T-cells (NFAT) is one of the main transcription factors driving the expression of inflammatory genes and thus regulating immune responses. NFAT-dependent production of inflammatory mediators is controlled by Ca2+-dependent protein phosphatase calcineurin (CaN), which dephosphorylates NFAT and promotes its transcriptional activity. A substantial body of evidence has demonstrated that aberrant CaN/NFAT signaling is linked to several pathologies observed in AD, including neuronal apoptosis, synaptic deficits, and glia activation. In view of this, the role of NFAT isoforms in AD has been linked to disease progression at different stages, some of which are paralleled to diminished cognitive status. The use of classical inhibitors of CaN/NFAT signaling, such as tacrolimus or cyclosporine, or adeno-associated viruses to specifically inhibit astrocytic NFAT activation, has alleviated some symptoms of AD by diminishing β-amyloid neurotoxicity and neuroinflammation. In this article, we discuss the recent findings related to the contribution of CaN/NFAT signaling to the progression of AD and highlight the possible benefits of targeting this pathway in AD treatment.
Collapse
Affiliation(s)
| | | | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
3
|
Hui W, Wenhua S, Shuojie Z, Lulin W, Panpan Z, Tongtong Z, Xiaoli X, Juhua D. How does NFAT3 regulate the occurrence of cardiac hypertrophy? IJC HEART & VASCULATURE 2023; 48:101271. [PMID: 37753338 PMCID: PMC10518445 DOI: 10.1016/j.ijcha.2023.101271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/24/2023] [Accepted: 09/08/2023] [Indexed: 09/28/2023]
Abstract
Cardiac hypertrophy is initially an adaptive response to physiological and pathological stimuli. Although pathological myocardial hypertrophy is the main cause of morbidity and mortality, our understanding of its mechanism is still weak. NFAT3 (nuclear factor of activated T-cell-3) is a member of the nuclear factor of the activated T cells (NFAT) family. NFAT3 plays a critical role in regulating the expression of cardiac hypertrophy genes by inducing their transcription. Recently, accumulating evidence has indicated that NFAT3 is a potent regulator of the progression of cardiac hypertrophy. This review, for the first time, summarizes the current studies on NFAT3 in cardiac hypertrophy, including the pathophysiological processes and the underlying pathological mechanism, focusing on the nuclear translocation and transcriptional function of NFAT3. This review will provide deep insight into the pathogenesis of cardiac hypertrophy and a theoretical basis for identifying new therapeutic targets in the NFAT3 network.
Collapse
Affiliation(s)
- Wang Hui
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Su Wenhua
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
- Department of Cardiology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Zhang Shuojie
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Wang Lulin
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Zhao Panpan
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Zhang Tongtong
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xie Xiaoli
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Dan Juhua
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
4
|
Lu Y, Liu M, Guo X, Wang P, Zeng F, Wang H, Tang J, Qin Z, Tao T. miR-26a-5p alleviates CFA-induced chronic inflammatory hyperalgesia through Wnt5a/CaMKII/NFAT signaling in mice. CNS Neurosci Ther 2023; 29:1254-1271. [PMID: 36756710 PMCID: PMC10068476 DOI: 10.1111/cns.14099] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Inflammation often leads to the occurrence of chronic pain, and many miRNAs have been shown to play a key role in the development of inflammatory pain. However, whether miR-26a-5p relieves pain induced by inflammation and its possible mechanism are still unclear. METHODS The complete Freund's adjuvant (CFA)-induced inflammatory pain mouse model was employed. Intrathecal or subcutaneous injection of miR-26a-5p agomir was performed after modeling to study its antinociceptive effect and the comparison of different administration methods. Bioinformatics analysis of miRNAs was performed to study the downstream mechanisms of miR-26a-5p. HE staining, RT-qPCR, Western blotting, and immunofluorescence were used for further validation. RESULTS A single intrathecal and subcutaneous injection of miR-26a-5p both reversed mechanical hypersensitivity and thermal latency in the left hind paw of mice with CFA-induced inflammatory pain. HE staining and immunofluorescence studies found that both administrations of miR-26a-5p alleviated inflammation in the periphery and spinal cord. Bioinformatics analysis and dual-luciferase reporter gene analysis identified Wnt5a as a direct downstream target gene of miR-26a-5p. Wnt5a was mainly expressed in neurons and microglia in the spinal cord of mice with inflammatory pain. Intrathecal injection of miR-26a-5p could significantly reduce the expression level of Wnt5a and inhibit the downstream molecules of noncanonical Wnt signaling Camk2/NFAT, inhibiting the release of spinal cord inflammatory factors and alleviating the activation of microglia. In addition, miR-26a-5p could also inhibit lipopolysaccharide (LPS)-stimulated BV2 cell inflammation in vitro through a noncanonical Wnt signaling pathway. CONCLUSIONS miR-26a-5p is a promising therapy for CFA-induced inflammatory pain. Both intrathecal and subcutaneous injections provide relief for inflammatory pain. miR-26a-5p regulated noncanonical Wnt signaling to be involved in analgesia partly through antineuroinflammation, suggesting a pain-alleviating effect via noncanonical Wnt signaling pathway in the CFA-induced inflammatory pain model in vivo.
Collapse
Affiliation(s)
- Yitian Lu
- Department of Anesthesiology, Nanfang hospital, Southern Medical University, Guangzhou, China.,Department of Anesthesiology, Central People's Hospital of Zhanjiang, Zhanjiang, China
| | - Maozhu Liu
- Department of pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangna Guo
- Department of Anesthesiology, Nanfang hospital, Southern Medical University, Guangzhou, China
| | - Peng Wang
- Department of Anesthesiology, Nanfang hospital, Southern Medical University, Guangzhou, China
| | - Fanning Zeng
- Department of Anesthesiology, Nanfang hospital, Southern Medical University, Guangzhou, China
| | - Haitao Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jing Tang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zaisheng Qin
- Department of Anesthesiology, Nanfang hospital, Southern Medical University, Guangzhou, China
| | - Tao Tao
- Department of Anesthesiology, Central People's Hospital of Zhanjiang, Zhanjiang, China
| |
Collapse
|
5
|
Arjun McKinney A, Petrova R, Panagiotakos G. Calcium and activity-dependent signaling in the developing cerebral cortex. Development 2022; 149:dev198853. [PMID: 36102617 PMCID: PMC9578689 DOI: 10.1242/dev.198853] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Calcium influx can be stimulated by various intra- and extracellular signals to set coordinated gene expression programs into motion. As such, the precise regulation of intracellular calcium represents a nexus between environmental cues and intrinsic genetic programs. Mounting genetic evidence points to a role for the deregulation of intracellular calcium signaling in neuropsychiatric disorders of developmental origin. These findings have prompted renewed enthusiasm for understanding the roles of calcium during normal and dysfunctional prenatal development. In this Review, we describe the fundamental mechanisms through which calcium is spatiotemporally regulated and directs early neurodevelopmental events. We also discuss unanswered questions about intracellular calcium regulation during the emergence of neurodevelopmental disease, and provide evidence that disruption of cell-specific calcium homeostasis and/or redeployment of developmental calcium signaling mechanisms may contribute to adult neurological disorders. We propose that understanding the normal developmental events that build the nervous system will rely on gaining insights into cell type-specific calcium signaling mechanisms. Such an understanding will enable therapeutic strategies targeting calcium-dependent mechanisms to mitigate disease.
Collapse
Affiliation(s)
- Arpana Arjun McKinney
- Graduate Program in Developmental and Stem Cell Biology, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA 94143, USA
| | - Ralitsa Petrova
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA 94143, USA
| | - Georgia Panagiotakos
- Graduate Program in Developmental and Stem Cell Biology, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
6
|
Jiang B, Ding T, Guo C, Bai X, Cao D, Wu X, Sha W, Jiang M, Wu L, Gao Y. NFAT1 Orchestrates Spinal Microglial Transcription and Promotes Microglial Proliferation via c-MYC Contributing to Nerve Injury-Induced Neuropathic Pain. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201300. [PMID: 35892263 PMCID: PMC9507349 DOI: 10.1002/advs.202201300] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 06/17/2022] [Indexed: 05/17/2023]
Abstract
Peripheral nerve injury-induced spinal microglial proliferation plays a pivotal role in neuropathic pain. So far, key intracellular druggable molecules involved in this process are not identified. The nuclear factor of activated T-cells (NFAT1) is a master regulator of immune cell proliferation. Whether and how NFAT1 modulates spinal microglial proliferation during neuropathic pain remain unknown. Here it is reported that NFAT1 is persistently upregulated in microglia after spinal nerve ligation (SNL), which is regulated by TET2-mediated DNA demethylation. Global or microglia-specific deletion of Nfat1 attenuates SNL-induced pain and decreases excitatory synaptic transmission of lamina II neurons. Furthermore, deletion of Nfat1 decreases microglial proliferation and the expression of multiple microglia-related genes, such as cytokines, transmembrane signaling receptors, and transcription factors. Particularly, SNL increases the binding of NFAT1 with the promoter of Itgam, Tnf, Il-1b, and c-Myc in the spinal cord. Microglia-specific overexpression of c-MYC induces pain hypersensitivity and microglial proliferation. Finally, inhibiting NFAT1 and c-MYC by intrathecal injection of inhibitor or siRNA alleviates SNL-induced neuropathic pain. Collectively, NFAT1 is a hub transcription factor that regulates microglial proliferation via c-MYC and guides the expression of the activated microglia genome. Thus, NFAT1 may be an effective target for treating neuropathic pain.
Collapse
Affiliation(s)
- Bao‐Chun Jiang
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Ting‐Yu Ding
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Chang‐Yun Guo
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Xue‐Hui Bai
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - De‐Li Cao
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Xiao‐Bo Wu
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Wei‐Lin Sha
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Ming Jiang
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Long‐Jun Wu
- Department of NeurologyMayo ClinicRochesterMN55905USA
| | - Yong‐Jing Gao
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| |
Collapse
|
7
|
Fu Y, Lorrai I, Zorman B, Mercatelli D, Shankula C, Marquez Gaytan J, Lefebvre C, de Guglielmo G, Kim HR, Sumazin P, Giorgi FM, Repunte-Canonigo V, Sanna PP. Escalated (Dependent) Oxycodone Self-Administration Is Associated with Cognitive Impairment and Transcriptional Evidence of Neurodegeneration in Human Immunodeficiency Virus (HIV) Transgenic Rats. Viruses 2022; 14:669. [PMID: 35458399 PMCID: PMC9030762 DOI: 10.3390/v14040669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/02/2022] [Accepted: 03/09/2022] [Indexed: 02/05/2023] Open
Abstract
Substance use disorder is associated with accelerated disease progression in people with human immunodeficiency virus (HIV; PWH). Problem opioid use, including high-dose opioid therapy, prescription drug misuse, and opioid abuse, is high and increasing in the PWH population. Oxycodone is a broadly prescribed opioid in both the general population and PWH. Here, we allowed HIV transgenic (Tg) rats and wildtype (WT) littermates to intravenously self-administer oxycodone under short-access (ShA) conditions, which led to moderate, stable, "recreational"-like levels of drug intake, or under long-access (LgA) conditions, which led to escalated (dependent) drug intake. HIV Tg rats with histories of oxycodone self-administration under LgA conditions exhibited significant impairment in memory performance in the novel object recognition (NOR) paradigm. RNA-sequencing expression profiling of the medial prefrontal cortex (mPFC) in HIV Tg rats that self-administered oxycodone under ShA conditions exhibited greater transcriptional evidence of inflammation than WT rats that self-administered oxycodone under the same conditions. HIV Tg rats that self-administered oxycodone under LgA conditions exhibited transcriptional evidence of an increase in neuronal injury and neurodegeneration compared with WT rats under the same conditions. Gene expression analysis indicated that glucocorticoid-dependent adaptations contributed to the gene expression effects of oxycodone self-administration. Overall, the present results indicate that a history of opioid intake promotes neuroinflammation and glucocorticoid dysregulation, and excessive opioid intake is associated with neurotoxicity and cognitive impairment in HIV Tg rats.
Collapse
Affiliation(s)
- Yu Fu
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, San Diego, CA 92037, USA; (Y.F.); (I.L.); (C.S.); (J.M.G.); (C.L.)
- European Bioinformatics Institute (EMBL-EBI), Hinxton CB10 1SD, UK
| | - Irene Lorrai
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, San Diego, CA 92037, USA; (Y.F.); (I.L.); (C.S.); (J.M.G.); (C.L.)
| | - Barry Zorman
- Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; (B.Z.); (H.R.K.); (P.S.)
| | - Daniele Mercatelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (D.M.); (F.M.G.)
| | - Chase Shankula
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, San Diego, CA 92037, USA; (Y.F.); (I.L.); (C.S.); (J.M.G.); (C.L.)
| | - Jorge Marquez Gaytan
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, San Diego, CA 92037, USA; (Y.F.); (I.L.); (C.S.); (J.M.G.); (C.L.)
| | - Celine Lefebvre
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, San Diego, CA 92037, USA; (Y.F.); (I.L.); (C.S.); (J.M.G.); (C.L.)
- 92160 Antony, France
| | - Giordano de Guglielmo
- Department of Psychiatry, University of California, La Jolla, San Diego, CA 92093, USA;
| | - Hyunjae Ryan Kim
- Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; (B.Z.); (H.R.K.); (P.S.)
| | - Pavel Sumazin
- Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; (B.Z.); (H.R.K.); (P.S.)
| | - Federico M. Giorgi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (D.M.); (F.M.G.)
| | - Vez Repunte-Canonigo
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, San Diego, CA 92037, USA; (Y.F.); (I.L.); (C.S.); (J.M.G.); (C.L.)
| | - Pietro Paolo Sanna
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, San Diego, CA 92037, USA; (Y.F.); (I.L.); (C.S.); (J.M.G.); (C.L.)
| |
Collapse
|
8
|
Basu-Shrivastava M, Kozoriz A, Desagher S, Lassot I. To Ubiquitinate or Not to Ubiquitinate: TRIM17 in Cell Life and Death. Cells 2021; 10:1235. [PMID: 34069831 PMCID: PMC8157266 DOI: 10.3390/cells10051235] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/17/2022] Open
Abstract
TRIM17 is a member of the TRIM family, a large class of RING-containing E3 ubiquitin-ligases. It is expressed at low levels in adult tissues, except in testis and in some brain regions. However, it can be highly induced in stress conditions which makes it a putative stress sensor required for the triggering of key cellular responses. As most TRIM members, TRIM17 can act as an E3 ubiquitin-ligase and promote the degradation by the proteasome of substrates such as the antiapoptotic protein MCL1. Intriguingly, TRIM17 can also prevent the ubiquitination of other proteins and stabilize them, by binding to other TRIM proteins and inhibiting their E3 ubiquitin-ligase activity. This duality of action confers several pivotal roles to TRIM17 in crucial cellular processes such as apoptosis, autophagy or cell division, but also in pathological conditions as diverse as Parkinson's disease or cancer. Here, in addition to recent data that endorse this duality, we review what is currently known from public databases and the literature about TRIM17 gene regulation and expression, TRIM17 protein structure and interactions, as well as its involvement in cell physiology and human disorders.
Collapse
Affiliation(s)
| | - Alina Kozoriz
- Institut de Génétique Moléculaire de Montpellier, University Montpellier, CNRS, Montpellier, France
| | - Solange Desagher
- Institut de Génétique Moléculaire de Montpellier, University Montpellier, CNRS, Montpellier, France
| | - Iréna Lassot
- Institut de Génétique Moléculaire de Montpellier, University Montpellier, CNRS, Montpellier, France
| |
Collapse
|
9
|
Liu X, Gao S, Gao H, Jiang X, Wei Q. Mitochondrial Disruption Is Involved in the Effect of Nuclear Factor of Activated T cells, Cytoplasmic 4 on Aggravating Cardiomyocyte Hypertrophy. J Cardiovasc Pharmacol 2021; 77:557-569. [PMID: 33951694 DOI: 10.1097/fjc.0000000000000986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/14/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Nuclear factor of activated T cells, cytoplasmic 4 (NFATc4), a nuclear transcription factor, has been implicated in cardiac hypertrophy through the enhancement of hypertrophic gene expression. However, the role of NFATc4 in mitochondrial modulation is mostly unknown. The current study aimed to investigate the role of NFATc4 in regulating mitochondrial function during phenylephrine (PE)-induced cardiac hypertrophy. Our results showed that overexpression of NFATc4 aggravated the PE-induced decrease in mitochondrial genesis, membrane potential, and mitochondrial gene expression as well as impaired mitochondrial respiration. However, knockdown of NFATc4 relieved PE-induced perturbations in mitochondria and cardiomyocyte hypertrophy. Mechanistically, by activating phosphoinositide-dependent kinase 1 and promoting a combination of AKT and phosphoinositide-dependent kinase 1, phosphorylation and sequential acetylation of PGC-1α were aggravated by NFATc4 and suppressed the activity of PGC-1α. In conclusion, NFATc4-regulated factors were shown to be associated with mitochondrial function and exacerbated PE-induced mitochondrial dysfunction. These findings revealed new roles of NFATc4 in cardiac hypertrophy.
Collapse
Affiliation(s)
- Xueping Liu
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi, People's Republic of China ; and
| | - Si Gao
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi, People's Republic of China ; and
| | - Hui Gao
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Xudong Jiang
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi, People's Republic of China ; and
| | - Qiqiu Wei
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi, People's Republic of China ; and
| |
Collapse
|
10
|
Kitamura N, Kaminuma O. Isoform-Selective NFAT Inhibitor: Potential Usefulness and Development. Int J Mol Sci 2021; 22:2725. [PMID: 33800389 PMCID: PMC7962815 DOI: 10.3390/ijms22052725] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/30/2022] Open
Abstract
Nuclear factor of activated T cells (NFAT), which is the pharmacological target of immunosuppressants cyclosporine and tacrolimus, has been shown to play an important role not only in T cells (immune system), from which their name is derived, but also in many biological events. Therefore, functional and/or structural abnormalities of NFAT are linked to the pathogenesis of diseases in various organs. The NFAT protein family consists of five isoforms, and each isoform performs diverse functions and has unique expression patterns in the target tissues. This diversity has made it difficult to obtain ideal pharmacological output for immunosuppressants that inhibit the activity of almost all NFAT family members, causing serious and wide-ranging side effects. Moreover, it remains unclear whether isoform-selective NFAT regulation can be achieved by targeting the structural differences among NFAT isoforms and whether this strategy can lead to the development of better drugs than the existing ones. This review summarizes the role of the NFAT family members in biological events, including the development of various diseases, as well as the usefulness of and problems associated with NFAT-targeting therapies, including those dependent on current immunosuppressants. Finally, we propose a novel therapeutic strategy based on the molecular mechanisms that enable selective regulation of specific NFAT isoforms.
Collapse
Affiliation(s)
- Noriko Kitamura
- Laboratory of Allergy and Immunology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan;
| | - Osamu Kaminuma
- Laboratory of Allergy and Immunology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan;
- Department of Disease Model, Research Institute of Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
| |
Collapse
|
11
|
Zhang Y, Chen D, Zhao L, Li W, Ni Y, Chen Y, Li H. Nfatc4 Deficiency Attenuates Ototoxicity by Suppressing Tnf-Mediated Hair Cell Apoptosis in the Mouse Cochlea. Front Immunol 2019; 10:1660. [PMID: 31379853 PMCID: PMC6650568 DOI: 10.3389/fimmu.2019.01660] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/03/2019] [Indexed: 12/12/2022] Open
Abstract
The loss of sensory hair cells in the cochlea is the major cause of sensorineural hearing loss, and inflammatory processes and immune factors in response to cochlear damage have been shown to induce hair cell apoptosis. The expression and function of Nfatc4 in the cochlea remains unclear. In this study, we investigated the expression of Nfatc4 in the mouse cochlea and explored its function using Nfatc4−/− mice. We first showed that Nfatc4 was expressed in the cochlear hair cells. Cochlear hair cell development and hearing function were normal in Nfatc4−/− mice, suggesting that Nfatc4 is not critical for cochlear development. We then showed that when the hair cells were challenged by ototoxic drugs Nfatc4 was activated and translocated from the cytoplasm to the nucleus, and this was accompanied by increased expression of Tnf and its downstream targets and subsequent hair cell apoptosis. Finally, we demonstrated that Nfatc4-deficient hair cells showed lower sensitivity to damage induced by ototoxic drugs and noise exposure compared to wild type controls. The Tnf-mediated apoptosis pathway was attenuated in Nfatc4-deficient cochlear epithelium, and this might be the reason for the reduced sensitivity of Nfatc4-deficient hair cells to injury. These findings suggest that the amelioration of inflammation-mediated hair cell apoptosis by inhibition of Nfatc4 activation might have significant therapeutic value in preventing ototoxic drug or noise exposure-induced sensorineural hearing loss.
Collapse
Affiliation(s)
- Yanping Zhang
- State Key Laboratory of Medical Neurobiology, Department of Affiliated Eye and ENT Hospital, ENT Institute and Otorhinolaryngology, Institutes of Biomedical Sciences and the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Diyan Chen
- State Key Laboratory of Medical Neurobiology, Department of Affiliated Eye and ENT Hospital, ENT Institute and Otorhinolaryngology, Institutes of Biomedical Sciences and the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Liping Zhao
- State Key Laboratory of Medical Neurobiology, Department of Affiliated Eye and ENT Hospital, ENT Institute and Otorhinolaryngology, Institutes of Biomedical Sciences and the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Wen Li
- State Key Laboratory of Medical Neurobiology, Department of Affiliated Eye and ENT Hospital, ENT Institute and Otorhinolaryngology, Institutes of Biomedical Sciences and the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Yusu Ni
- State Key Laboratory of Medical Neurobiology, Department of Affiliated Eye and ENT Hospital, ENT Institute and Otorhinolaryngology, Institutes of Biomedical Sciences and the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Yan Chen
- State Key Laboratory of Medical Neurobiology, Department of Affiliated Eye and ENT Hospital, ENT Institute and Otorhinolaryngology, Institutes of Biomedical Sciences and the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Huawei Li
- State Key Laboratory of Medical Neurobiology, Department of Affiliated Eye and ENT Hospital, ENT Institute and Otorhinolaryngology, Institutes of Biomedical Sciences and the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China.,Shanghai Engineering Research Centre of Cochlear Implant, Shanghai, China
| |
Collapse
|
12
|
Mitochondrial calcium signalling and neurodegenerative diseases. Neuronal Signal 2018; 2:NS20180061. [PMID: 32714593 PMCID: PMC7373239 DOI: 10.1042/ns20180061] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/06/2018] [Accepted: 09/20/2018] [Indexed: 12/11/2022] Open
Abstract
Calcium is utilised by cells in signalling and in regulating ATP production; it also contributes to cell survival and, when concentrations are unbalanced, triggers pathways for cell death. Mitochondria contribute to calcium buffering, meaning that mitochondrial calcium uptake and release is intimately related to cytosolic calcium concentrations. This review focuses on the proteins contributing to mitochondrial calcium homoeostasis, the roles of the mitochondrial permeability transition pore (MPTP) and mitochondrial calcium-activated proteins, and their relevance in neurodegenerative pathologies. It also covers alterations to calcium homoeostasis in Friedreich ataxia (FA).
Collapse
|
13
|
Hwang E, Gao W, Xiao Y, Ngo HTT, Yi T. Helianthus annuus
L. flower prevents UVB‐induced photodamage in human dermal fibroblasts by regulating the MAPK/AP‐1, NFAT, and Nrf2 signaling pathways. J Cell Biochem 2018; 120:601-612. [DOI: 10.1002/jcb.27417] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/12/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Eunson Hwang
- Graduate School of Biotechnology, Kyung Hee University Giheung‐gu Yongin‐si Gyeonggi‐do South Korea
| | - Wei Gao
- Graduate School of Biotechnology, Kyung Hee University Giheung‐gu Yongin‐si Gyeonggi‐do South Korea
| | - Yong‐kun Xiao
- Graduate School of Biotechnology, Kyung Hee University Giheung‐gu Yongin‐si Gyeonggi‐do South Korea
- Tianjin Ginkingsen Health Technology Co., Ltd Binhai Gaoxin Tianjin China
| | - Hien T. T. Ngo
- Graduate School of Biotechnology, Kyung Hee University Giheung‐gu Yongin‐si Gyeonggi‐do South Korea
| | - Tae‐Hoo Yi
- Graduate School of Biotechnology, Kyung Hee University Giheung‐gu Yongin‐si Gyeonggi‐do South Korea
| |
Collapse
|
14
|
Tan Z, Kang T, Zhang X, Tong Y, Chen S. Nerve growth factor prevents arsenic-induced toxicity in PC12 cells through the AKT/GSK-3β/NFAT pathway. J Cell Physiol 2018; 234:4726-4738. [PMID: 30256405 DOI: 10.1002/jcp.27255] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/25/2018] [Indexed: 12/13/2022]
Abstract
The potential risk of arsenic-related neurodegeneration has been a growing concern. Arsenic exposure has been reported to disrupt neurite growth and neuron body integrity in vitro; however, its underlying mechanism remains unclear. Previously, we showed that arsenic sulfide (AS) exerted cytotoxicity in gastric and colon cancer cells through regulating nuclear factor of the activated T cells (NFAT) pathway. The NFAT pathway regulates axon path finding and neural development. Using neural crest cell line PC12 cells as a model, here we show that AS caused mitochondrial membrane potential collapse, reactive oxygen species production, and cytochrome c release, leading to mitochondria-mediated apoptosis via the AKT/GSK-3β/NFAT pathway. Increased glycogen synthase kinase-3 beta (GSK-3β) activation leads to the inactivation of NFAT and its antiapoptotic effects. Through inhibiting GSK-3β activity, both nerve growth factor (NGF) and Tideglusib, a GSK-3β inhibitor partially rescued the PC12 cells from the AS-induced cytotoxicity and restored the expression of NFATc3. In addition, overexpression of NFATc3 stimulated neurite outgrowth and potentiated the effect of NGF on promoting the neurite outgrowth. Collectively, our results show that NFATc3 serves as the downstream target of NGF and plays a key role in preventing AS-induced neurotoxicity through regulating the AKT/GSK-3β/NFAT pathway in PC12 cells.
Collapse
Affiliation(s)
- Zhen Tan
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Kang
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiuli Zhang
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yingying Tong
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Siyu Chen
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
15
|
Hopp SC, Bihlmeyer NA, Corradi JP, Vanderburg C, Cacace AM, Das S, Clark TW, Betensky RA, Hyman BT, Hudry E. Neuronal calcineurin transcriptional targets parallel changes observed in Alzheimer disease brain. J Neurochem 2018; 147:24-39. [PMID: 29806693 DOI: 10.1111/jnc.14469] [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] [Received: 02/26/2018] [Revised: 04/11/2018] [Accepted: 05/09/2018] [Indexed: 01/23/2023]
Abstract
Synaptic dysfunction and loss are core pathological features in Alzheimer disease (AD). In the vicinity of amyloid-β plaques in animal models, synaptic toxicity occurs and is associated with chronic activation of the phosphatase calcineurin (CN). Indeed, pharmacological inhibition of CN blocks amyloid-β synaptotoxicity. We therefore hypothesized that CN-mediated transcriptional changes may contribute to AD neuropathology and tested this by examining the impact of CN over-expression on neuronal gene expression in vivo. We found dramatic transcriptional down-regulation, especially of synaptic mRNAs, in neurons chronically exposed to CN activation. Importantly, the transcriptional profile parallels the changes in human AD tissue. Bioinformatics analyses suggest that both nuclear factor of activated T cells and numerous microRNAs may all be impacted by CN, and parallel findings are observed in AD. These data and analyses support the hypothesis that at least part of the synaptic failure characterizing AD may result from aberrant CN activation leading to down-regulation of synaptic genes, potentially via activation of specific transcription factors and expression of repressive microRNAs. OPEN PRACTICES Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/ Read the Editorial Highlight for this article on page 8.
Collapse
Affiliation(s)
- Sarah C Hopp
- Alzheimer's disease Research Laboratory, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Nathan A Bihlmeyer
- MIND Informatics, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Cambridge, Massachusetts, USA
| | - John P Corradi
- Exploratory Biology and Genomics, Bristol-Myers Squibb, Wallingford, Connecticut, USA
| | - Charles Vanderburg
- Alzheimer's disease Research Laboratory, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Angela M Cacace
- Exploratory Biology and Genomics, Bristol-Myers Squibb, Wallingford, Connecticut, USA
| | - Sudeshna Das
- MIND Informatics, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Cambridge, Massachusetts, USA
| | - Timothy W Clark
- MIND Informatics, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Cambridge, Massachusetts, USA
| | - Rebecca A Betensky
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Bradley T Hyman
- Alzheimer's disease Research Laboratory, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Eloise Hudry
- Alzheimer's disease Research Laboratory, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| |
Collapse
|
16
|
Zhao QR, Lu JM, Li ZY, Mei YA. Neuritin promotes neurite and spine growth in rat cerebellar granule cells via L-type calcium channel-mediated calcium influx. J Neurochem 2018; 147:40-57. [PMID: 29920676 PMCID: PMC6220818 DOI: 10.1111/jnc.14535] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/21/2018] [Accepted: 06/13/2018] [Indexed: 01/15/2023]
Abstract
Neuritin is a neurotrophic factor that is activated by neural activity and neurotrophins. Its major function is to promote neurite growth and branching; however, the underlying mechanisms are not fully understood. To address this issue, this study investigated the effects of neuritin on neurite and spine growth and intracellular Ca2+ concentration in rat cerebellar granule neurons (CGNs). Incubation of CGNs for 24 h with neuritin increased neurite length and spine density; this effect was mimicked by insulin and abolished by inhibiting insulin receptor (IR) or mitogen‐activated protein kinase kinase/extracellular signal‐regulated kinase (ERK) activity. Calcium imaging and western blot analysis revealed that neuritin enhanced the increase in intracellular Ca2+ level induced by high K+, and stimulated the cell surface expression of CaV1.2 and CaV1.3 α subunits of the L‐type calcium channel, which was suppressed by inhibition of IR or mitogen‐activated protein kinase kinase/ERK. Treatment with inhibitors of L‐type calcium channels, calmodulin, and calcineurin (CaN) abrogated the effects of neuritin on neurite length and spine density. A similar result was obtained by silencing nuclear factor of activated T cells c4, which is known to be activated by neuritin in CGNs. These results indicate that IR and ERK signaling as well as the Ca2+/CaN/nuclear factor of activated T cells c4 axis mediate the effects of neuritin on neurite and spine growth in CGNs. Open Practices
Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/ ![]()
Cover Image for this issue: doi: 10.1111/jnc.14195.
Collapse
Affiliation(s)
- Qian-Ru Zhao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, China
| | - Jun-Mei Lu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, China
| | - Zhao-Yang Li
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, China
| | - Yan-Ai Mei
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Ding B, Dobner PR, Mullikin-Kilpatrick D, Wang W, Zhu H, Chow CW, Cave JW, Gronostajski RM, Kilpatrick DL. BDNF activates an NFI-dependent neurodevelopmental timing program by sequestering NFATc4. Mol Biol Cell 2018; 29:975-987. [PMID: 29467254 PMCID: PMC5896935 DOI: 10.1091/mbc.e16-08-0595] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 02/07/2018] [Accepted: 02/13/2018] [Indexed: 12/20/2022] Open
Abstract
We show that BDNF regulates the timing of neurodevelopment via a novel mechanism of extranuclear sequestration of NFATc4 in Golgi. This leads to accelerated derepression of an NFI temporal occupancy gene program in cerebellar granule cells that includes Bdnf itself, revealing an autoregulatory loop within the program driven by BDNF and NFATc4.
Collapse
Affiliation(s)
- Baojin Ding
- Department of Microbiology and Physiological Systems and Program in Neuroscience, University of Massachusetts Medical School, Worcester, MA 01605-2324
| | - Paul R. Dobner
- Department of Microbiology and Physiological Systems and Program in Neuroscience, University of Massachusetts Medical School, Worcester, MA 01605-2324
| | - Debra Mullikin-Kilpatrick
- Department of Microbiology and Physiological Systems and Program in Neuroscience, University of Massachusetts Medical School, Worcester, MA 01605-2324
| | - Wei Wang
- Department of Microbiology and Physiological Systems and Program in Neuroscience, University of Massachusetts Medical School, Worcester, MA 01605-2324
| | - Hong Zhu
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10461
| | - Chi-Wing Chow
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10461
| | - John W. Cave
- Burke Medical Research Institute, White Plains, NY 10605
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065
| | - Richard M. Gronostajski
- Department of Biochemistry, Program in Neuroscience and Developmental Genomics Group, New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY 14203
| | - Daniel L. Kilpatrick
- Department of Microbiology and Physiological Systems and Program in Neuroscience, University of Massachusetts Medical School, Worcester, MA 01605-2324
| |
Collapse
|
18
|
Purroy R, Britti E, Delaspre F, Tamarit J, Ros J. Mitochondrial pore opening and loss of Ca 2+ exchanger NCLX levels occur after frataxin depletion. Biochim Biophys Acta Mol Basis Dis 2018; 1864:618-631. [PMID: 29223733 DOI: 10.1016/j.bbadis.2017.12.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/29/2017] [Accepted: 12/05/2017] [Indexed: 12/12/2022]
Abstract
Frataxin-deficient neonatal rat cardiomyocytes and dorsal root ganglia neurons have been used as cell models of Friedreich ataxia. In previous work we show that frataxin depletion resulted in mitochondrial swelling and lipid droplet accumulation in cardiomyocytes, and compromised DRG neurons survival. Now, we show that these cells display reduced levels of the mitochondrial calcium transporter NCLX that can be restored by calcium-chelating agents and by external addition of frataxin fused to TAT peptide. Also, the transcription factor NFAT3, involved in cardiac hypertrophy and apoptosis, becomes activated by dephosphorylation in both cardiomyocytes and DRG neurons. In cardiomyocytes, frataxin depletion also results in mitochondrial permeability transition pore opening. Since the pore opening can be inhibited by cyclosporin A, we show that this treatment reduces lipid droplets and mitochondrial swelling in cardiomyocytes, restores DRG neuron survival and inhibits NFAT dephosphorylation. These results highlight the importance of calcium homeostasis and that targeting mitochondrial pore by repurposing cyclosporin A, could be envisaged as a new strategy to treat the disease.
Collapse
Affiliation(s)
- R Purroy
- Department of Ciències Mèdiques Bàsiques, Fac. Medicina, University of Lleida, IRB Lleida, Lleida, Spain
| | - E Britti
- Department of Ciències Mèdiques Bàsiques, Fac. Medicina, University of Lleida, IRB Lleida, Lleida, Spain
| | - F Delaspre
- Department of Ciències Mèdiques Bàsiques, Fac. Medicina, University of Lleida, IRB Lleida, Lleida, Spain
| | - J Tamarit
- Department of Ciències Mèdiques Bàsiques, Fac. Medicina, University of Lleida, IRB Lleida, Lleida, Spain
| | - J Ros
- Department of Ciències Mèdiques Bàsiques, Fac. Medicina, University of Lleida, IRB Lleida, Lleida, Spain.
| |
Collapse
|
19
|
Kaminuma O, Kitamura N, Nishito Y, Nemoto S, Tatsumi H, Mori A, Hiroi T. Downregulation of NFAT3 Due to Lack of T-Box Transcription Factor TBX5 Is Crucial for Cytokine Expression in T Cells. THE JOURNAL OF IMMUNOLOGY 2017; 200:92-100. [PMID: 29180489 DOI: 10.4049/jimmunol.1602113] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 10/19/2017] [Indexed: 11/19/2022]
Abstract
The NFAT family transcription factors play crucial roles in immunological and other biological activities. NFAT3 is rarely expressed in T cells, and the mechanisms and significance of the specific NFAT3 downregulation in T cells have been unknown. In human CD4+ T cells, overexpression of NFAT1 and NFAT3 enhanced and suppressed IL-2 expression, respectively. NFAT3 downregulation in Jurkat cells using RNA interference technology augmented IL-2 expression, whereas a knockdown of NFAT1, NFAT2, and NFAT4 suppressed it. The promoter/enhancer activity of the NFAT-binding site in the IL-2 gene was upregulated and downregulated by NFAT1 and NFAT3, respectively. A study employing NFAT1/NFAT3 chimeric molecules revealed that the region in NFAT3 responsible for NFAT promoter activity inhibition was located within its N-terminal transactivation domain, Ca2+-regulatory domain, and DNA-binding domain. Downregulation of NFAT3 expression in T cells is mediated by lower chromatin accessibility and enhancer activity in its promoter in comparison with aortic smooth muscle cells expressing endogenous NFAT3. The binding sites of T-box transcription factor TBX5 and NK-2 transcription factor-related locus 5 Nkx2.5, which were expressed at higher levels in aortic smooth muscle cells than in T cells, were located within the -387 to +97 NFAT3 promoter region, exhibiting the maximum enhancer activity. Mutating the binding site of TBX5 but not Nkx2.5 diminished the NFAT3 promoter activity, whereas the overexpression of TBX5 enhanced it. Introduction of TBX5 into CD4+ T cells enhanced the expression of NFAT3 and suppressed that of IL-2. TBX5 deficiency-mediated downregulation of NFAT3 is crucial for the high cytokine-producing activity of T cells.
Collapse
Affiliation(s)
- Osamu Kaminuma
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo 113-8613, Japan; .,Center for Life Science Research, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Noriko Kitamura
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo 113-8613, Japan
| | - Yasumasa Nishito
- Center for Basic Technology Research, The Tokyo Metropolitan Institute of Medical Science, Tokyo 113-8613, Japan
| | - Soichi Nemoto
- Department of Obstetrics and Gynecology, National Hospital Organization, Sagamihara National Hospital, Kanagawa 252-0392, Japan; and
| | - Hideki Tatsumi
- Department of Obstetrics and Gynecology, National Hospital Organization, Sagamihara National Hospital, Kanagawa 252-0392, Japan; and
| | - Akio Mori
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization, Sagamihara National Hospital, Kanagawa 252-0392, Japan
| | - Takachika Hiroi
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo 113-8613, Japan
| |
Collapse
|
20
|
Rahimian R, Béland LC, Kriz J. Galectin-3: mediator of microglia responses in injured brain. Drug Discov Today 2017; 23:375-381. [PMID: 29133191 DOI: 10.1016/j.drudis.2017.11.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 10/23/2017] [Accepted: 11/07/2017] [Indexed: 12/15/2022]
Abstract
Galectin-3 is a pleiotropic protein involved in cell activation, proliferation and migration and plays a pivotal part as an inflammatory mediator in neurodegeneration. Galectin-3 is associated with microglial activation and proliferation after ischemia. Given its putative role as a dynamic fine-tuner of microglia, activation of Galectin-3 provides molecular cues in design of new immunomodulatory strategies for stroke management. This review summarizes recent evidence on the role of Galectin-3 as a mediator of immune responses in damaged brain and mechanisms employed by Galectin-3 to affect microglial function.
Collapse
Affiliation(s)
- Reza Rahimian
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Center, Laval University, Quebec, Quebec G1J 2G3, Canada
| | - Louis-Charles Béland
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Center, Laval University, Quebec, Quebec G1J 2G3, Canada
| | - Jasna Kriz
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Center, Laval University, Quebec, Quebec G1J 2G3, Canada; Faculty of Medicine, Department of Psychiatry and Neuroscience, Laval University, Québec, Québec, Canada.
| |
Collapse
|
21
|
Xu M, Fan Q, Zhang J, Chen Y, Xu R, Chen L, Zhao P, Tian Y. NFAT3/c4-mediated excitotoxicity in hippocampal apoptosis during radiation-induced brain injury. JOURNAL OF RADIATION RESEARCH 2017; 58:827-833. [PMID: 28992110 PMCID: PMC5710526 DOI: 10.1093/jrr/rrx041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Indexed: 05/09/2023]
Abstract
Whole brain irradiation (WBI) has become an indispensible tool in the treatment of head and neck cancer, and it has greatly improved patient survival rate and total survival time. In addition, prophylactic cranial irradiation (PCI) has dramatically decreased the incidence of brain metastatic carcinoma. However, WBI may induce temporary functional deficits or even progressive, irreversible cognitive dysfunction that compromises the quality of life for survivors. Unfortunately, the exact molecular mechanisms for cognitive damage remain elusive, and no treatment or preventative measures are available for use in the clinic. In the present study, the nuclear factor of activated T cells isoform 4 (NFAT3/c4) was found to play a vital role in excitotoxic hippocampus cell apoptosis induced by radiation. Sprague-Dawley (SD) rats received 20 Gy WBI, after which we detected NFAT3/c4-mediated excitotoxicity. We found that radiation caused hippocampus excitotoxicity, resulting from overactivation of the N-methyl-D-aspartate receptor (NMDAR) and always accompanied by subsequent elevation of the intracellular calcium level and activation of calcineurin (CaN). P-NFAT3/c4 was the principal downstream target of CaN, including regulation of its nuclear translocation as well as transcriptional activities. Radiation recruited NMDAR/NFAT3/c4 activation and subsequent Bax induction in hippocampus cells. Once treated with the NFAT3/c4 inhibitor 11R-VIVIT peptide pre-irradiation, hippocampal proliferation and neuron survival (dentate gyrus cells in particular) were protected from radiation-induced injury, resulting in inhibition of the apoptosis marker Bax. Our principal aim was to illuminate the role of NFAT3/c4-mediated excitotoxicity in hippocampal apoptosis during radiation-induced brain injury. This study is the first time that radiation-induced activation of NFAT3/c4 has been recorded, and our results suggest that NFAT3/c4 may be a novel target for prevention and treatment of radiation-induced brain injury.
Collapse
Affiliation(s)
- Meiling Xu
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou, Jiangsu 215004, China
| | - Qiuhong Fan
- Institute of Radiotherapy & Oncology, Soochow University
| | - Junjun Zhang
- Suzhou Key Laboratory for Radiation Oncology, San Xiang Road No. 1055, Suzhou 215004, China
| | - Yanfang Chen
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou, Jiangsu 215004, China
| | - Ruizhe Xu
- Institute of Radiotherapy & Oncology, Soochow University
| | - Liesong Chen
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou, Jiangsu 215004, China
| | - Peifeng Zhao
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou, Jiangsu 215004, China
| | - Ye Tian
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou, Jiangsu 215004, China
- Corresponding author. Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou, Jiangsu 215004, China. Tel.: +86-512-6778-3430; Fax: +86-512-6828-4303; E-mail:
| |
Collapse
|
22
|
Perroud J, Bernheim L, Frieden M, Koenig S. Distinct roles of NFATc1 and NFATc4 in human primary myoblast differentiation and in the maintenance of reserve cells. J Cell Sci 2017; 130:3083-3093. [PMID: 28760926 DOI: 10.1242/jcs.198978] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 07/25/2017] [Indexed: 01/06/2023] Open
Abstract
Ca2+ signaling plays a key role during human myoblast differentiation. Among Ca2+-sensitive pathways, calcineurin is essential for myoblast differentiation and muscle regeneration. Nuclear factor of activated T-cell (NFAT) transcription factors are the major calcineurin targets. We investigated the expression and the role of each NFAT gene during human primary myoblast differentiation. We found that three NFAT isoforms are present, NFATc1, NFATc3 and NFATc4. Importantly, while their mRNA expression increases during differentiation, NFATc1 is more highly expressed in myotubes, whilst NFATc4 is specifically maintained in reserve cells. NFATc3 is present in both cell types, although no specific role during myoblast differentiation was observed. Knockdown of either NFATc1 or NFATc4 affects the differentiation process similarly, by decreasing the expression of late differentiation markers, but impairs myotube formation differently. Whereas NFATc1 knockdown strongly reduced the number and the surface area of myotubes, NFATc4 knockdown increased the surface area of myotubes and reduced the pool of reserve cells. We conclude that NFAT genes have specific roles in myotube formation and in the maintenance of the reserve cell pool during human postnatal myogenesis.
Collapse
Affiliation(s)
- Julie Perroud
- Department of Basic Neurosciences, University Medical Center, Rue Michel Servet 1, 1211 Geneva 4, Switzerland
| | - Laurent Bernheim
- Department of Basic Neurosciences, University Medical Center, Rue Michel Servet 1, 1211 Geneva 4, Switzerland
| | - Maud Frieden
- Department of Cell Physiology and Metabolism, University Medical Center, Rue Michel Servet 1, 1211 Geneva 4, Switzerland
| | - Stephane Koenig
- Department of Basic Neurosciences, University Medical Center, Rue Michel Servet 1, 1211 Geneva 4, Switzerland
| |
Collapse
|
23
|
Tozaki-Saitoh H, Miyata H, Yamashita T, Matsushita K, Tsuda M, Inoue K. P2Y12 receptors in primary microglia activate nuclear factor of activated T-cell signaling to induce C-C chemokine 3 expression. J Neurochem 2017; 141:100-110. [PMID: 28144998 DOI: 10.1111/jnc.13968] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/16/2017] [Accepted: 01/18/2017] [Indexed: 02/03/2023]
Abstract
Microglia are widely accepted as surveillants in the central nervous system that are continually searching the local environment for signs of injury. Following an inflammatory situation, microglia alter their morphology, extend ramified processes, and undergo cell body hypertrophy. Extracellular nucleotides are recognized as a danger signal by microglia. ADP acting on P2Y12 receptors induce process extension of microglia thereby attracting microglia to the site of adenosine tri-phosphate/ADP leaking or release. However, the question whether ADP/P2Y12 receptor signaling directly stimulates the production or release of inducible factors such as cytokines remains unclear. In this study, we found that CC chemokine ligand 3 (CCL3) is induced by ADP-treated primary microglia. Pharmacological characterization using pertussis toxin, a P2Y12 receptor inhibitor, and a calcium chelator revealed that CCL3 induction was caused by P2Y12 receptor-mediated intracellular calcium elevation. Next, nuclear factor of activated T-cell dephosphorylation and nuclear translocalization were observed. Calcineurin, an inhibitor for nuclear factor of activated T cell, suppressed CCL3 induction. These data indicate that microglial P2Y12 receptors are utilized to trigger an acute inflammatory response in microglia via rapid CCL3 induction after ADP stimulation.
Collapse
Affiliation(s)
- Hidetoshi Tozaki-Saitoh
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.,Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Miyata
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomohiro Yamashita
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsuyuki Matsushita
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu Universisty, Fukuoka, Japan
| | - Makoto Tsuda
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuhide Inoue
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
24
|
Boczek T, Lisek M, Ferenc B, Zylinska L. Cross talk among PMCA, calcineurin and NFAT transcription factors in control of calmodulin gene expression in differentiating PC12 cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:502-515. [PMID: 28153703 DOI: 10.1016/j.bbagrm.2017.01.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/16/2017] [Accepted: 01/27/2017] [Indexed: 11/19/2022]
Abstract
Brain aging is characterized by progressive loss of plasma membrane calcium pump (PMCA) and its activator - calmodulin (CaM), but the mechanism of this phenomenon remains unresolved. CaM encoded by three genes Calm1, Calm2, Calm3, works to translate Ca2+ signal into changes in frequently opposite cellular activities. This unique function allows CaM to affect gene expression via stimulation of calcineurin (CaN) and its downstream target - nuclear factor of activated T-cells (NFAT) and to terminate Ca2+ signal by stimulation of its extrusion. PMCA, which exists in four isoforms PMCA1-4, may in turn shape the pattern of Ca2+ transients and control CaN activity by its direct binding. Therefore, the interplay between PMCA, CaM and CaN/NFAT is highly plausible. To verify that, we used differentiated PC12 cells with reduced expression of PMCA2 or PMCA3 to mimic the potential changes in aged brain. Manipulation in PMCAs level decreased CaM protein in PMCA2 or PMCA3-reduced lines that was accompanied by down-regulation of Calm1 and Calm2 in both lines, but Calm3 only in PMCA2-reduced cells. Further studies showed substantially higher NFATc2 nuclear accumulation and increased NFAT transcriptional activity. Blocking of CaN/NFAT signalling resulted in almost full CaM recovery, mainly due to up-regulation of Calm2 and Calm3 genes. Moreover, higher occupancy of Calm2 and Calm3 promoters by NFATc2 and increased expression of these genes in response to NFATc2 silencing were demonstrated in PMCA2 and PMCA3-reduced lines. Our results indicate that decrease in CaM level in response to PMCAs downregulation can be driven by CaN/NFAT pathway.
Collapse
Affiliation(s)
- Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University, Mazowiecka 6/8 Str., 92-215 Lodz, Poland; Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA.
| | - Malwina Lisek
- Department of Molecular Neurochemistry, Medical University, Mazowiecka 6/8 Str., 92-215 Lodz, Poland
| | - Bozena Ferenc
- Department of Molecular Neurochemistry, Medical University, Mazowiecka 6/8 Str., 92-215 Lodz, Poland
| | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Medical University, Mazowiecka 6/8 Str., 92-215 Lodz, Poland
| |
Collapse
|
25
|
Manocha GD, Ghatak A, Puig KL, Kraner SD, Norris CM, Combs CK. NFATc2 Modulates Microglial Activation in the AβPP/PS1 Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2017; 58:775-787. [PMID: 28505967 PMCID: PMC6265241 DOI: 10.3233/jad-151203] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) brains are characterized by fibrillar amyloid-β (Aβ) peptide containing plaques and associated reactive microglia. The proinflammatory phenotype of the microglia suggests that they may negatively affect disease course and contribute to behavioral decline. This hypothesis predicts that attenuating microglial activation may provide benefit against disease. Prior work from our laboratory and others has characterized a role for the transcription factor, nuclear factor of activated T cells (NFAT), in regulating microglial phenotype in response to different stimuli, including Aβ peptide. We observed that the NFATc2 isoform was the most highly expressed in murine microglia cultures, and inhibition or deletion of NFATc2 was sufficient to attenuate the ability of the microglia to secrete cytokines. In order to determine whether the NFATc2 isoform, in particular, was a valid immunomodulatory target in vivo, we crossed an NFATc2-/- line to a well-known AD mouse model, an AβPP/PS1 mouse line. As expected, the AβPP/PS1 x NFATc2-/- mice had attenuated cytokine levels compared to AβPP/PS1 mice as well as reduced microgliosis and astrogliosis with no effect on plaque load. Although some species differences in relative isoform expression may exist between murine and human microglia, it appears that microglial NFAT activity is a viable target for modulating the proinflammatory changes that occur during AD.
Collapse
Affiliation(s)
- Gunjan D. Manocha
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Atreyi Ghatak
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Kendra L. Puig
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Susan D. Kraner
- Department of Pharmacology and Nutritional Sciences and the Sanders Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Christopher M. Norris
- Department of Pharmacology and Nutritional Sciences and the Sanders Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Colin K. Combs
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| |
Collapse
|
26
|
Bodmer D, Perkovic A, Sekulic-Jablanovic M, Wright MB, Petkovic V. Pasireotide prevents nuclear factor of activated T cells nuclear translocation and acts as a protective agent in aminoglycoside-induced auditory hair cell loss. J Neurochem 2016; 139:1113-1123. [PMID: 27787949 DOI: 10.1111/jnc.13880] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 10/18/2016] [Accepted: 10/19/2016] [Indexed: 12/23/2022]
Abstract
Hearing impairment is a global health problem with a high socioeconomic impact. Damage to auditory hair cells (HCs) in the inner ear as a result of aging, disease, trauma, or toxicity, underlies the majority of cases of sensorineural hearing loss. Previously we demonstrated that the Ca2+ -sensitive neuropeptide, somatostatin (SST), and an analog, octreotide, protect HCs from gentamicin-induced cell death in vitro. Aminoglycosides such as gentamicin trigger a calcium ion influx (Ca2+ ) that activates pro-apoptotic signaling cascades in HCs. SST binding to the G-protein-coupled receptors (SSTR1-SSTR5) that are directly linked to voltage-dependent Ca2+ channels inhibits Ca2+ channel activity and associated downstream events. Here, we report that the SST analog pasireotide, a high affinity ligand to SSTRs 1-3, and 5, with a longer half-life than octreotide, prevents gentamicin-induced HC death in the mouse organ of Corti (OC). Explant experiments using OCs derived from SSTR1 and SSTR1and 2 knockout mice, revealed that SSTR2 mediates pasireotide's anti-apoptotic effects. Mechanistically, pasireotide prevented a nuclear translocation of the Ca2+ -sensitive transcription factor, nuclear factor of activated T cells (NFAT), which is ordinarily provoked by gentamicin in OC explants. Direct inhibition of NFAT with 11R-VIVIT also prevented the gentamicin-dependent nuclear translocation of NFAT and apoptosis. Both pasireotide and 11R-VIVIT partially reversed the effects of gentamicin on the expression of downstream survival targets (NMDA receptor and the regulatory subunit of phosphatidylinositol-4,5-bisphosphate 3-kinase, PI3K). These data suggest that SST analogs antagonize aminoglycoside-induced cell death in an NFAT-dependent fashion. SST analogs and NFAT inhibitors may therefore offer new therapeutic possibilities for the treatment of hearing loss.
Collapse
Affiliation(s)
- Daniel Bodmer
- Department of Biomedicine and the Clinic for Otorhinolaryngology, University Hospital Basel, Basel, Switzerland
| | - Adrijana Perkovic
- Department of Biomedicine and the Clinic for Otorhinolaryngology, University Hospital Basel, Basel, Switzerland
| | - Marijana Sekulic-Jablanovic
- Department of Biomedicine and the Clinic for Otorhinolaryngology, University Hospital Basel, Basel, Switzerland
| | | | - Vesna Petkovic
- Department of Biomedicine and the Clinic for Otorhinolaryngology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
27
|
Yao JJ, Zhao QR, Liu DD, Chow CW, Mei YA. Neuritin Up-regulates Kv4.2 α-Subunit of Potassium Channel Expression and Affects Neuronal Excitability by Regulating the Calcium-Calcineurin-NFATc4 Signaling Pathway. J Biol Chem 2016; 291:17369-81. [PMID: 27307045 PMCID: PMC5016134 DOI: 10.1074/jbc.m115.708883] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Indexed: 11/06/2022] Open
Abstract
Neuritin is an important neurotrophin that regulates neural development, synaptic plasticity, and neuronal survival. Elucidating the downstream molecular signaling is important for potential therapeutic applications of neuritin in neuronal dysfunctions. We previously showed that neuritin up-regulates transient potassium outward current (IA) subunit Kv4.2 expression and increases IA densities, in part by activating the insulin receptor signaling pathway. Molecular mechanisms of neuritin-induced Kv4.2 expression remain elusive. Here, we report that the Ca(2+)/calcineurin (CaN)/nuclear factor of activated T-cells (NFAT) c4 axis is required for neuritin-induced Kv4.2 transcriptional expression and potentiation of IA densities in cerebellum granule neurons. We found that neuritin elevates intracellular Ca(2+) and increases Kv4.2 expression and IA densities; this effect was sensitive to CaN inhibition and was eliminated in Nfatc4(-/-) mice but not in Nfatc2(-/-) mice. Stimulation with neuritin significantly increased nuclear accumulation of NFATc4 in cerebellum granule cells and HeLa cells, which expressed IR. Furthermore, NFATc4 was recruited to the Kv4.2 gene promoter loci detected by luciferase reporter and chromatin immunoprecipitation assays. More importantly, data obtained from cortical neurons following adeno-associated virus-mediated overexpression of neuritin indicated that reduced neuronal excitability and increased formation of dendritic spines were abrogated in the Nfatc4(-/-) mice. Together, these data demonstrate an indispensable role for the CaN/NFATc4 signaling pathway in neuritin-regulated neuronal functions.
Collapse
Affiliation(s)
- Jin-Jing Yao
- From the Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China and
| | - Qian-Ru Zhao
- From the Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China and
| | - Dong-Dong Liu
- From the Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China and
| | - Chi-Wing Chow
- the Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York 10461
| | - Yan-Ai Mei
- From the Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China and
| |
Collapse
|
28
|
Cell cycle and apoptosis regulation by NFAT transcription factors: new roles for an old player. Cell Death Dis 2016; 7:e2199. [PMID: 27100893 PMCID: PMC4855676 DOI: 10.1038/cddis.2016.97] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/13/2016] [Accepted: 03/16/2016] [Indexed: 12/11/2022]
Abstract
The NFAT (nuclear factor of activated T cells) family of transcription factors consists of four Ca2+-regulated members (NFAT1–NFAT4), which were first described in T lymphocytes. In addition to their well-documented role in T lymphocytes, where they control gene expression during cell activation and differentiation, NFAT proteins are also expressed in a wide range of cells and tissue types and regulate genes involved in cell cycle, apoptosis, angiogenesis and metastasis. The NFAT proteins share a highly conserved DNA-binding domain (DBD), which allows all NFAT members to bind to the same DNA sequence in enhancers or promoter regions. The same DNA-binding specificity suggests redundant roles for the NFAT proteins, which is true during the regulation of some genes such as IL-2 and p21. However, it has become increasingly clear that different NFAT proteins and even isoforms can have unique functions. In this review, we address the possible reasons for these distinct roles, particularly regarding N- and C-terminal transactivation regions (TADs) and the partner proteins that interact with these TADs. We also discuss the genes regulated by NFAT during cell cycle regulation and apoptosis and the role of NFAT during tumorigenesis.
Collapse
|
29
|
Toth AB, Shum AK, Prakriya M. Regulation of neurogenesis by calcium signaling. Cell Calcium 2016; 59:124-34. [PMID: 27020657 DOI: 10.1016/j.ceca.2016.02.011] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 12/22/2022]
Abstract
Calcium (Ca(2+)) signaling has essential roles in the development of the nervous system from neural induction to the proliferation, migration, and differentiation of neural cells. Ca(2+) signaling pathways are shaped by interactions among metabotropic signaling cascades, intracellular Ca(2+) stores, ion channels, and a multitude of downstream effector proteins that activate specific genetic programs. The temporal and spatial dynamics of Ca(2+) signals are widely presumed to control the highly diverse yet specific genetic programs that establish the complex structures of the adult nervous system. Progress in the last two decades has led to significant advances in our understanding of the functional architecture of Ca(2+) signaling networks involved in neurogenesis. In this review, we assess the literature on the molecular and functional organization of Ca(2+) signaling networks in the developing nervous system and its impact on neural induction, gene expression, proliferation, migration, and differentiation. Particular emphasis is placed on the growing evidence for the involvement of store-operated Ca(2+) release-activated Ca(2+) (CRAC) channels in these processes.
Collapse
Affiliation(s)
- Anna B Toth
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Andrew K Shum
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Murali Prakriya
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, United States.
| |
Collapse
|
30
|
Vihma H, Luhakooder M, Pruunsild P, Timmusk T. Regulation of different human NFAT isoforms by neuronal activity. J Neurochem 2016; 137:394-408. [PMID: 26851544 DOI: 10.1111/jnc.13568] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 01/04/2016] [Accepted: 01/29/2016] [Indexed: 12/30/2022]
Abstract
Nuclear factor of activated T-cells (NFAT) is a family of transcription factors comprising four calcium-regulated members: NFATc1, NFATc2, NFATc3, and NFATc4. Upon activation by the calcium-dependent phosphatase calcineurin (CaN), NFATs translocate from cytosol to the nucleus and regulate their target genes, which in the nervous system are involved in axon growth, synaptic plasticity, and neuronal survival. We have shown previously that there are a number of different splice variants of NFAT genes expressed in the brain. Here, we studied the subcellular localizations and transactivation capacities of alternative human NFAT isoforms in rat primary cortical or hippocampal neurons in response to membrane depolarization and compared the induced transactivation levels in neurons to those obtained from HEK293 cells in response to calcium signaling. We confirm that in neurons the translocation to the nucleus of all NFAT isoforms is reliant on the activity of CaN. However, our results suggest that both the regulation of subcellular localization and transcriptional activity of NFAT proteins in neurons is isoform specific. We show that in primary hippocampal neurons NFATc2 isoforms have very fast translocation kinetics, whereas NFATc4 isoforms translocate relatively slowly to the nucleus. Moreover, we demonstrate that the strongest transcriptional activators in HEK293 cells are NFATc1 and NFATc3, but in neurons NFATc3 and NFATc4 lead to the highest induction, and NFATc2 and NFATc1 display isoform-specific transcription activation capacities. Altogether, our results indicate that the effects of calcium signaling on the action of NFAT proteins are isoform-specific and can differ between cell types. We show that the effects of calcium signaling on the action of NFAT proteins are isoform-specific and differ between cell types. Although nuclear localization of all NFAT isoforms in neurons requires calcineurin, the subcellular distributions, neuronal activity-induced nuclear translocation extent and kinetics, and transcription activation capacities of alternative NFAT proteins vary.
Collapse
Affiliation(s)
- Hanna Vihma
- Department of Gene Technology, Tallinn University of Technology, Tallinn, Estonia
| | - Mirjam Luhakooder
- Department of Gene Technology, Tallinn University of Technology, Tallinn, Estonia
| | - Priit Pruunsild
- Department of Gene Technology, Tallinn University of Technology, Tallinn, Estonia
| | - Tõnis Timmusk
- Department of Gene Technology, Tallinn University of Technology, Tallinn, Estonia
| |
Collapse
|
31
|
Sawamura S, Hatano M, Takada Y, Hino K, Kawamura T, Tanikawa J, Nakagawa H, Hase H, Nakao A, Hirano M, Rotrattanadumrong R, Kiyonaka S, Mori MX, Nishida M, Hu Y, Inoue R, Nagata R, Mori Y. Screening of Transient Receptor Potential Canonical Channel Activators Identifies Novel Neurotrophic Piperazine Compounds. Mol Pharmacol 2016; 89:348-63. [PMID: 26733543 DOI: 10.1124/mol.115.102863] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/04/2016] [Indexed: 12/23/2022] Open
Abstract
Transient receptor potential canonical (TRPC) proteins form Ca(2+)-permeable cation channels activated upon stimulation of metabotropic receptors coupled to phospholipase C. Among the TRPC subfamily, TRPC3 and TRPC6 channels activated directly by diacylglycerol (DAG) play important roles in brain-derived neurotrophic factor (BDNF) signaling, promoting neuronal development and survival. In various disease models, BDNF restores neurologic deficits, but its therapeutic potential is limited by its poor pharmacokinetic profile. Elucidation of a framework for designing small molecules, which elicit BDNF-like activity via TRPC3 and TRPC6, establishes a solid basis to overcome this limitation. We discovered, through library screening, a group of piperazine-derived compounds that activate DAG-activated TRPC3/TRPC6/TRPC7 channels. The compounds [4-(5-chloro-2-methylphenyl)piperazin-1-yl](3-fluorophenyl)methanone (PPZ1) and 2-[4-(2,3-dimethylphenyl)piperazin-1-yl]-N-(2-ethoxyphenyl)acetamide (PPZ2) activated, in a dose-dependent manner, recombinant TRPC3/TRPC6/TRPC7 channels, but not other TRPCs, in human embryonic kidney cells. PPZ2 activated native TRPC6-like channels in smooth muscle cells isolated from rabbit portal vein. Also, PPZ2 evoked cation currents and Ca(2+) influx in rat cultured central neurons. Strikingly, both compounds induced BDNF-like neurite growth and neuroprotection, which were abolished by a knockdown or inhibition of TRPC3/TRPC6/TRPC7 in cultured neurons. Inhibitors of Ca(2+) signaling pathways, except calcineurin, impaired neurite outgrowth promotion induced by PPZ compounds. PPZ2 increased activation of the Ca(2+)-dependent transcription factor, cAMP response element-binding protein. These findings suggest that Ca(2+) signaling mediated by activation of DAG-activated TRPC channels underlies neurotrophic effects of PPZ compounds. Thus, piperazine-derived activators of DAG-activated TRPC channels provide important insights for future development of a new class of synthetic neurotrophic drugs.
Collapse
Affiliation(s)
- Seishiro Sawamura
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Masahiko Hatano
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Yoshinori Takada
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Kyosuke Hino
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Tetsuya Kawamura
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Jun Tanikawa
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Hiroshi Nakagawa
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Hideharu Hase
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Akito Nakao
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Mitsuru Hirano
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Rachapun Rotrattanadumrong
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Shigeki Kiyonaka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Masayuki X Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Motohiro Nishida
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Yaopeng Hu
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Ryuji Inoue
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Ryu Nagata
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering (S.S., Ma.H., Y.T., H.H., Mi.H., R.R., S.K., M.X.M., Y.M.), and Department of Technology and Ecology, Hall of Global Environmental Studies (S.K., Y.M.), Kyoto University, Kyoto, Japan; Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan (Y.T., K.H., T.K., J.T., H.N., R.N.); Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan (A.N.); Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan (M.N.); and Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (Y.H., R.I.)
| |
Collapse
|
32
|
Toll-Like Receptors Promote Mitochondrial Translocation of Nuclear Transcription Factor Nuclear Factor of Activated T-Cells in Prolonged Microglial Activation. J Neurosci 2015. [PMID: 26224862 DOI: 10.1523/jneurosci.2455-14.2015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Microglia are resident macrophages in the CNS that scavenge pathogens, dying cells, and molecules using pattern recognition Toll-like receptors (TLRs). Nuclear factor of activated T-cells (NFAT) family transcription factors also regulate inflammatory responses in microglia. However, whether there exists cross talk between TLR and NFAT signaling is unclear. Here we show that chronic activation of murine microglia by prolonged stimulation of Toll-like receptor 4 (TLR4) ligand lipopolysaccharides (LPSs) leads to unexpected translocation of NFAT1 into mitochondria. This mitochondrial import of NFAT1 is independent of calcium/calcineurin signaling. Instead, inhibition of Toll/interleukin 1 receptor domain-containing adapter-inducing interferon-β (TRIF) pathway blocks the mitochondrial translocation of NFAT1. Functionally, inhibition of NFAT1 reduces the TRIF-mediated expression of interferon-β and compromises the production of ATP and reactive oxygen species in LPS-treated microglia. Therefore, our findings reveal a new inflammatory signaling pathway that links TLR with NFAT in regulating cytokine production and mitochondrial activity during chronic microglial activation. SIGNIFICANCE STATEMENT Nuclear factor of activated T-cells (NFAT) family transcription factors are known to undergo nuclear translocation in response to inflammatory stimulation. In this study, we uncovered a surprise transportation of NFATs into mitochondria in microglia after a prolonged treatment with bacteria endotoxin lipopolysaccharides (LPSs). LPSs activated Toll-like receptor 4 and its downstream Toll/interleukin 1 receptor-domain-containing adapter-inducing interferon-β (TRIF) to regulate the mitochondrial translocation of NFAT in microglia, whereas genetic inhibition of NFAT1 compromised TRIF-mediated cytokine production and reduced ATP and reactive oxygen species generation. These findings reveal a previously undescribed mitochondrial translocation of NFAT in microglia responding to extended activation of Toll-like receptor-mediated signaling transduction pathways.
Collapse
|
33
|
Moreno M, Fernández V, Monllau JM, Borrell V, Lerin C, de la Iglesia N. Transcriptional Profiling of Hypoxic Neural Stem Cells Identifies Calcineurin-NFATc4 Signaling as a Major Regulator of Neural Stem Cell Biology. Stem Cell Reports 2015; 5:157-65. [PMID: 26235896 PMCID: PMC4618660 DOI: 10.1016/j.stemcr.2015.06.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 06/29/2015] [Accepted: 06/29/2015] [Indexed: 01/09/2023] Open
Abstract
Neural stem cells (NSCs) reside in a hypoxic microenvironment within the brain. However, the crucial transcription factors (TFs) that regulate NSC biology under physiologic hypoxia are poorly understood. Here we have performed gene set enrichment analysis (GSEA) of microarray datasets from hypoxic versus normoxic NSCs with the aim of identifying pathways and TFs that are activated under oxygen concentrations mimicking normal brain tissue microenvironment. Integration of TF target (TFT) and pathway enrichment analysis identified the calcium-regulated TF NFATc4 as a major candidate to regulate hypoxic NSC functions. Nfatc4 expression was coordinately upregulated by top hypoxia-activated TFs, while NFATc4 target genes were enriched in hypoxic NSCs. Loss-of-function analyses further revealed that the calcineurin-NFATc4 signaling axis acts as a major regulator of NSC self-renewal and proliferation in vitro and in vivo by promoting the expression of TFs, including Id2, that contribute to the maintenance of the NSC state.
Collapse
Affiliation(s)
- Marta Moreno
- Clinical and Experimental Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Virginia Fernández
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Spain
| | - Josep M Monllau
- Clinical and Experimental Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Spain
| | - Carles Lerin
- Endocrinology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Núria de la Iglesia
- Clinical and Experimental Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.
| |
Collapse
|
34
|
Rojanathammanee L, Floden AM, Manocha GD, Combs CK. Attenuation of microglial activation in a mouse model of Alzheimer's disease via NFAT inhibition. J Neuroinflammation 2015; 12:42. [PMID: 25889879 PMCID: PMC4355356 DOI: 10.1186/s12974-015-0255-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/25/2015] [Indexed: 12/21/2022] Open
Abstract
Background Amyloid β (Aβ) peptide is hypothesized to stimulate microglia to acquire their characteristic proinflammatory phenotype in Alzheimer’s disease (AD) brains. The specific mechanisms by which Aβ leads to microglial activation remain an area of interest for identifying attractive molecular targets for intervention. Based upon the fact that microglia express the proinflammatory transcription factor, nuclear factor of activated T cells (NFAT), we hypothesized that NFAT activity is required for the Aβ-stimulated microgliosis that occurs during disease. Methods Primary murine microglia cultures were stimulated with Aβ in the absence or presence of NFAT inhibitors, FK506 and tat-VIVIT peptide, to quantify secretion of cytokines, neurotoxins, or Aβ phagocytosis. A transgenic mouse model of AD, APP/PS1, was treated subcutaneously via mini-osmotic pumps with FK506 or tat-VIVIT to quantify effects on cytokines, microgliosis, plaque load, and memory. Results Expression of various NFAT isoforms was verified in primary murine microglia through Western blot analysis. Microglial cultures were stimulated with Aβ fibrils in the absence or presence of the NFAT inhibitors, FK506 and tat-VIVIT, to demonstrate that NFAT activity regulated Aβ phagocytosis, neurotoxin secretion, and cytokine secretion. Delivery of FK506 and tat-VIVIT to transgenic APP/PS1 mice attenuated spleen but not brain cytokine levels. However, FK506 and tat-VIVIT significantly attenuated both microgliosis and Aβ plaque load in treated mice compared to controls. Surprisingly, this did not correlate with changes in memory performance via T-maze testing. Conclusions Our findings suggest that development of specific NFAT inhibitors may offer promise as an effective strategy for attenuating the microgliosis and Aβ plaque deposition that occur in AD. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0255-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lalida Rojanathammanee
- Institute of Science, Suranaree University of Technology, 111 University Avenue, Suranaree Subdistric, Nakhon Ratchasima, 30000, Thailand. .,Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, 504 Hamline Street, Neuroscience Building, Grand Forks, ND, 58203, USA.
| | - Angela M Floden
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, 504 Hamline Street, Neuroscience Building, Grand Forks, ND, 58203, USA.
| | - Gunjan D Manocha
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, 504 Hamline Street, Neuroscience Building, Grand Forks, ND, 58203, USA.
| | - Colin K Combs
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, 504 Hamline Street, Neuroscience Building, Grand Forks, ND, 58203, USA.
| |
Collapse
|
35
|
Serrano-Pérez MC, Fernández M, Neria F, Berjón-Otero M, Doncel-Pérez E, Cano E, Tranque P. NFAT transcription factors regulate survival, proliferation, migration, and differentiation of neural precursor cells. Glia 2015; 63:987-1004. [DOI: 10.1002/glia.22797] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 01/08/2015] [Accepted: 01/09/2015] [Indexed: 01/16/2023]
Affiliation(s)
- María C. Serrano-Pérez
- Laboratorio de Neuroglía, Instituto de Investigación en Discapacidades Neurológicas (IDINE); Universidad de Castilla-La Mancha (UCLM); Albacete Spain
| | - Miriam Fernández
- Laboratorio de Neuroglía, Instituto de Investigación en Discapacidades Neurológicas (IDINE); Universidad de Castilla-La Mancha (UCLM); Albacete Spain
| | - Fernando Neria
- Unidad de Neuroinflamación, Unidad Funcional de Investigaciones en Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII); Madrid Spain
| | - Mónica Berjón-Otero
- Laboratorio de Neuroglía, Instituto de Investigación en Discapacidades Neurológicas (IDINE); Universidad de Castilla-La Mancha (UCLM); Albacete Spain
| | - Ernesto Doncel-Pérez
- Grupo de Química Neuro-regenerativa, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla La Mancha (SESCAM); Toledo Spain
| | - Eva Cano
- Unidad de Neuroinflamación, Unidad Funcional de Investigaciones en Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII); Madrid Spain
| | - Pedro Tranque
- Laboratorio de Neuroglía, Instituto de Investigación en Discapacidades Neurológicas (IDINE); Universidad de Castilla-La Mancha (UCLM); Albacete Spain
| |
Collapse
|
36
|
Kim MS, Shutov LP, Gnanasekaran A, Lin Z, Rysted JE, Ulrich JD, Usachev YM. Nerve growth factor (NGF) regulates activity of nuclear factor of activated T-cells (NFAT) in neurons via the phosphatidylinositol 3-kinase (PI3K)-Akt-glycogen synthase kinase 3β (GSK3β) pathway. J Biol Chem 2014; 289:31349-60. [PMID: 25231981 DOI: 10.1074/jbc.m114.587188] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The Ca(2+)/calcineurin-dependent transcription factor nuclear factor of activated T-cells (NFAT) plays an important role in regulating many neuronal functions, including excitability, axonal growth, synaptogenesis, and neuronal survival. NFAT can be activated by action potential firing or depolarization that leads to Ca(2+)/calcineurin-dependent dephosphorylation of NFAT and its translocation to the nucleus. Recent data suggest that NFAT and NFAT-dependent functions in neurons can also be potently regulated by NGF and other neurotrophins. However, the mechanisms of NFAT regulation by neurotrophins are not well understood. Here, we show that in dorsal root ganglion sensory neurons, NGF markedly facilitates NFAT-mediated gene expression induced by mild depolarization. The effects of NGF were not associated with changes in [Ca(2+)]i and were independent of phospholipase C activity. Instead, the facilitatory effect of NGF depended on activation of the PI3K/Akt pathway downstream of the TrkA receptor and on inhibition of glycogen synthase kinase 3β (GSK3β), a protein kinase known to phosphorylate NFAT and promote its nuclear export. Knockdown or knockout of NFATc3 eliminated this facilitatory effect. Simultaneous monitoring of EGFP-NFATc3 nuclear translocation and [Ca(2+)]i changes in dorsal root ganglion neurons indicated that NGF slowed the rate of NFATc3 nuclear export but did not affect its nuclear import rate. Collectively, our data suggest that NGF facilitates depolarization-induced NFAT activation by stimulating PI3K/Akt signaling, inactivating GSK3β, and thereby slowing NFATc3 export from the nucleus. We propose that NFAT serves as an integrator of neurotrophin action and depolarization-driven calcium signaling to regulate neuronal gene expression.
Collapse
Affiliation(s)
- Man-Su Kim
- From the Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and the College of Pharmacy, Inje University, Gimhae 621-749, Korea
| | - Leonid P Shutov
- From the Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and
| | - Aswini Gnanasekaran
- From the Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and
| | - Zhihong Lin
- From the Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and
| | - Jacob E Rysted
- From the Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and
| | - Jason D Ulrich
- From the Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and
| | - Yuriy M Usachev
- From the Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and
| |
Collapse
|
37
|
Control of neuronal apoptosis by reciprocal regulation of NFATc3 and Trim17. Cell Death Differ 2014; 22:274-86. [PMID: 25215946 DOI: 10.1038/cdd.2014.141] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 07/08/2014] [Accepted: 08/04/2014] [Indexed: 12/18/2022] Open
Abstract
Neuronal apoptosis induced by survival factor deprivation is strongly regulated at the transcriptional level. Notably, the nuclear factor of activated T cell (NFAT) transcription factors have an important role in the control of the survival/death fate of neurons. However, the mechanisms that regulate NFAT activity in response to apoptotic stimuli and the target genes that mediate their effect on neuronal apoptosis are mostly unknown. In a previous study, we identified Trim17 as a crucial E3 ubiquitin ligase that is necessary and sufficient for neuronal apoptosis. Here, we show that Trim17 binds preferentially SUMOylated forms of NFATc3. Nonetheless, Trim17 does not promote the ubiquitination/degradation of NFATc3. NFAT transcription factors are regulated by calcium/calcineurin-dependent nuclear-cytoplasmic shuttling. Interestingly, Trim17 reduced by twofold the calcium-mediated nuclear localization of NFATc3 and, consistent with this, halved NFATc3 activity, as estimated by luciferase assays and by measurement of target gene expression. Trim17 also inhibited NFATc4 nuclear translocation and activity. NFATc4 is known to induce the expression of survival factors and, as expected, overexpression of NFATc4 protected cerebellar granule neurons from serum/KCl deprivation-induced apoptosis. Inhibition of NFATc4 by Trim17 may thus partially mediate the proapoptotic effect of Trim17. In contrast, overexpression of NFATc3 aggravated neuronal death, whereas knockdown of NFATc3 protected neurons from apoptosis. This proapoptotic effect of NFATc3 might be due to a feedback loop in which NFATc3, but not NFATc4, induces the transcription of the proapoptotic gene Trim17. Indeed, we found that overexpression or silencing of NFATc3, respectively, increased or decreased Trim17 levels, whereas NFATc4 had no significant effect on Trim17 expression. Moreover, we showed that NFATc3 binds to the promoter of the Trim17 gene together with c-Jun. Therefore, our results describe a novel mechanism regulating NFAT transcription factors beyond the calcium/calcineurin-dependent pathway and provide a possible explanation for the opposite effects of NFATc3 and NFATc4 on neuronal apoptosis.
Collapse
|
38
|
Zhang L, Li R, Shi W, Liang X, Liu S, Ye Z, Yu C, Chen Y, Zhang B, Wang W, Lai Y, Ma J, Li Z, Tan X. NFAT2 inhibitor ameliorates diabetic nephropathy and podocyte injury in db/db mice. Br J Pharmacol 2014; 170:426-39. [PMID: 23826864 DOI: 10.1111/bph.12292] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 06/18/2013] [Accepted: 06/21/2013] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND PURPOSE Podocyte injury plays a key role in the development of diabetic nephropathy (DN). We have recently shown that 11R-VIVIT, an inhibitor of cell-permeable nuclear factor of activated T-cells (NFAT), attenuates podocyte apoptosis induced by high glucose in vitro. However, it is not known whether 11R-VIVIT has a protective effect on DN, especially podocyte injury, under in vivo diabetic conditions. Hence, we examined the renoprotective effects of 11R-VIVIT in diabetic db/db mice and the possible mechanisms underlying its protective effects on podocyte injury in vivo and in vitro. EXPERIMENTAL APPROACH Type 2 diabetic db/db mice received i.p. injections of 11R-VIVIT (1 mg·kg(-1)) three times a week and were killed after 8 weeks. Immortalized mouse podocytes were cultured under different experimental conditions. KEY RESULTS 11R-VIVIT treatment markedly attenuated the albuminuria in diabetic db/db mice and also alleviated mesangial matrix expansion and podocyte injury. However, body weight, food and water intake, and glucose levels were unaffected. It also attenuated the increased NFAT2 activation and enhanced urokinase-type plasminogen activator receptor (uPA receptor) expression in glomerulor podocytes. In cultured podocytes, the increased nuclear accumulation of NFAT2 and uPA receptor expression induced by high glucose treatment was prevented by 11R-VIVIT or NFAT2-knockdown; this was accompanied by improvements in the filtration barrier function of the podocyte monolayer. CONCLUSIONS AND IMPLICATIONS The NFAT inhibitor 11R-VIVIT might be a useful therapeutic strategy for protecting podocytes and treating DN. The calcinerin/NFAT2/uPA receptor signalling pathway should be exploited as a therapeutic target for protecting podocytes from injury in DN.
Collapse
Affiliation(s)
- Li Zhang
- Southern Medical University, Guangzhou, China; Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Heat shock factor 1 (HSF1) protects neurons from death caused by the accumulation of misfolded proteins. It is believed that this protective effect is mediated by the transcriptional stimulation of genes encoding heat shock proteins (HSPs), a family of chaperones that refold or degrade misfolded proteins. Whether HSF1 is protective when neuronal death is not caused by protein misfolding has not been studied. Here, we report that HSF1 expression is necessary for the survival of rat neurons and that HSF1 mRNA and protein expression is reduced in neurons primed to die. Knock-down of HSF1 induces death of otherwise healthy neurons, whereas reestablishment of elevated levels of HSF1 protects neurons even when death is not due to accumulation of misfolded proteins. Neuroprotection by HSF1 does not require its trimerization, an event obligatory for the binding of HSF1 to heat shock elements within HSP gene promoters. Moreover, knock-down of HSP70 or blockade of HSP90 signaling does not reduce neuroprotection by HSF1. Although several neuroprotective molecules and signaling pathways, including CaMK, PKA, Casein kinase-II, and the Raf-MEK-ERK and PI-3K-Akt pathways, are not required for HSF1-mediated neuroprotection, protection is abrogated by inhibition of classical histone deacetylases (HDACs). We report that the novel mechanism of neuroprotection by HSF1 involves cooperation with SIRT1, an HDAC with well documented neuroprotective effects. Using a cell culture model of Huntington's disease, we show that HSF1 trimerization is not required for protection against mutant huntingtin-induced neurotoxicity, suggesting that HSF1 can protect neurons against both proteinopathic and nonproteinopathic death through a noncanonical pathway.
Collapse
|
40
|
Kosiorek M, Zylinska L, Zablocki K, Pikula S. Calcineurin/NFAT signaling represses genes Vamp1 and Vamp2 via PMCA-dependent mechanism during dopamine secretion by Pheochromocytoma cells. PLoS One 2014; 9:e92176. [PMID: 24667359 PMCID: PMC3965406 DOI: 10.1371/journal.pone.0092176] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 02/19/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Plasma membrane Ca(2+)-ATPases (PMCA) extrude Ca(2+) ions out of the cell and contribute to generation of calcium oscillations. Calcium signaling is crucial for transcriptional regulation of dopamine secretion by neuroendocrine PC12 cells. Low resting [Ca(2+)]c in PC12 cells is maintained mainly by two Ca(2+)-ATPases, PMCA2 and PMCA3. Recently, we found that Ca(2+) dependent phosphatase calcineurin was excessively activated under conditions of experimental downregulation of PMCA2 or PMCA3. Thus, the aim of this study was to explain if, via modulation of the Ca(2+)/calcineurin-dependent nuclear factor of activated T cells (NFAT) pathway, PMCA2 and PMCA3 affect intracellular signaling in pheochromocytoma/neuronal cells/PC12 cells. Secondly, we tested whether this might influence dopamine secretion by PC12 cells. RESULTS PMCA2- and PMCA3-deficient cells displayed profound decrease in dopamine secretion accompanied by a permanent increase in [Ca(2+)]c. Reduction in secretion might result from changes in NFAT signaling, following altered PMCA pattern. Consequently, activation of NFAT1 and NFAT3 transcription factors was observed in PMCA2- or PMCA3-deficient cells. Furthermore, chromatin immunoprecipitation assay indicated that NFATs could be involved in repression of Vamp genes encoding vesicle associated membrane proteins (VAMP). CONCLUSIONS PMCA2 and PMCA3 are crucial for dopamine secretion in PC12 cells. Reduction in PMCA2 or PMCA3 led to calcium-dependent activation of calcineurin/NFAT signaling and, in consequence, to repression of the Vamp gene and deterioration of the SNARE complex formation in PC12 cells.
Collapse
Affiliation(s)
- Michalina Kosiorek
- Department of Biochemistry, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
- Laboratory of Neurogenetics, Department of Neurodegenerative Disorders, Mossakowski Medical Research Centre PAS, Warsaw, Poland
- * E-mail: (MK); (SP)
| | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Medical University, Lodz, Poland
| | - Krzysztof Zablocki
- Department of Biochemistry, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| | - Slawomir Pikula
- Department of Biochemistry, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
- * E-mail: (MK); (SP)
| |
Collapse
|
41
|
Yan HQ, Shin SS, Ma X, Li Y, Dixon CE. Differential effect of traumatic brain injury on the nuclear factor of activated T Cells C3 and C4 isoforms in the rat hippocampus. Brain Res 2013; 1548:63-72. [PMID: 24389074 DOI: 10.1016/j.brainres.2013.12.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 12/20/2013] [Accepted: 12/23/2013] [Indexed: 10/25/2022]
Abstract
The interaction between the phosphatase calcineurin and transcription factor nuclear factor of activated T cells (NFAT) plays an important role numerous signaling and the regulatory events. Although NFAT is mostly known for its transcription function in the immune system, NFAT also has essential functions even in the central nervous system (CNS). The effects of traumatic brain injury (TBI) on NFAT are currently unknown. To determine if there is an alteration in NFAT after TBI, we examined NFATc3 and c4 levels at 6 h, 1 day, 1 week, 2 weeks and 4 weeks post injury. Rats were anesthetized and surgically prepared for controlled cortical impact (CCI) injury or sham surgery. Semi-quantitative measurements of NFATc3 and c4 in the hippocampal homogenates from injured and sham rats sacrificed at the appropriate time after injury were assessed using Western blot analysis. After TBI insult, in the hippocampus ipsilateral to the injury, NFATc3 expression levels were decreased both in the cytoplasmic and nuclear fractions. However, NFATc4 expression levels were increased in the cytoplasmic fraction but decreased in the nuclear fraction. Double labeling (with NeuN and GFAP) immunohistochemistry revealed that NFATc3 was expressed in subset of astrocytes and NFATc4 was expressed primarily in neurons. These differential responses in NFATc3 and c4 expression after TBI insult may indicate long-term changes in hippocampal excitability and may contribute to behavioral deficits. Further study is warranted to illustrate the role of NFATc3 and c4 in the setting of TBI.
Collapse
Affiliation(s)
- Hong Q Yan
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Samuel S Shin
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Xiecheng Ma
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Youming Li
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - C Edward Dixon
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15260.,Veterans Affairs Pittsburgh Healthcare System Pittsburgh, PA 15240
| |
Collapse
|
42
|
Spatiotemporal changes in NFATc4 expression of retinal ganglion cells after light-induced damage. J Mol Neurosci 2013; 53:69-77. [PMID: 24362677 DOI: 10.1007/s12031-013-0198-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 11/28/2013] [Indexed: 12/27/2022]
Abstract
Nuclear factor of activated T cells, cytoplasmic 4 (NFATc4) is one of the four members of the NFAT family, which were described first as essential components of T cells activation and lately as important regulators for the initiation and coordination of the immune response, including B cells and natural killer cells. Accumulating evidence has demonstrated that NFATc4 exerted a pro-apoptotic effect in the pathogenesis of various experimental central nervous system diseases by upregulating Fas ligand (FasL) levels. However, the function of NFATc4 in the retina is still with limited acquaintance. To investigate whether NFATc4 is involved in retinal neuron apoptosis, we performed a light-induced retinal damage model in adult rats. A significant upregulation of NFATc4 was detected in the retina after light-induced damage by using Western blotting and reverse transcriptase PCR (RT-PCR). Besides this, NFATc4 was observed to be localized mainly in the retinal ganglion cells (RGCs). In addition, the expression patterns of active caspase-3, active caspase-8, and FasL were parallel with that of NFATc4. We also found the co-localization of NFATc4 with active caspase-3 and FasL in RGCs after light exposure. Collectively, we hypothesized that NFATc4 might participate in RGCs apoptosis by upregulating FasL levels.
Collapse
|
43
|
Stefos GC, Soppa U, Dierssen M, Becker W. NGF upregulates the plasminogen activation inhibitor-1 in neurons via the calcineurin/NFAT pathway and the Down syndrome-related proteins DYRK1A and RCAN1 attenuate this effect. PLoS One 2013; 8:e67470. [PMID: 23825664 PMCID: PMC3692457 DOI: 10.1371/journal.pone.0067470] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 05/18/2013] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Plasminogen activator inhibitor 1 (PAI-1) is a key regulator of the plasminogen activation system. Although several lines of evidence support a significant role of PAI-1 in the brain, the regulation of its expression in neurons is poorly understood. In the present study we tested the hypothesis that NGF induces the upregulation of PAI-1 via the calcineurin/nuclear factor of activated T cells (NFAT) pathway and analysed whether the overexpression of the Down syndrome-related proteins DYRK1A and RCAN1 modulated the effect of NGF on PAI-1 expression. RESULTS NGF upregulated PAI-1 mRNA levels in primary mouse hippocampal neurons cultured for 3 days in vitro and in the rat pheochromocytoma cell line PC12. Reporter gene assays revealed that NGF activated the calcineurin/NFAT pathway in PC12 cells. Induction of PAI-1 by NGF was sensitive to the calcineurin inhibitor FK506 and the specific inhibition of NFAT activation by the cell permeable VIVIT peptide. Activation of calcineurin/NFAT signalling through other stimuli resulted in a much weaker induction of PAI-1 expression, suggesting that other NGF-induced pathways are involved in PAI-1 upregulation. Overexpression of either DYRK1A or RCAN1 negatively regulated NFAT-dependent transcriptional activity and reduced the upregulation of PAI-1 levels by NGF. CONCLUSION The present results show that the calcineurin/NFAT pathway mediates the upregulation of PAI-1 by NGF. The negative effect of DYRK1A and RCAN1 overexpression on NGF signal transduction in neural cells may contribute to the altered neurodevelopment and brain function in Down syndrome.
Collapse
Affiliation(s)
- Georgios C Stefos
- Institute of Pharmacology and Toxicology, Medical Faculty of the RWTH Aachen University, Aachen, Germany.
| | | | | | | |
Collapse
|
44
|
Murase S. Signal transducer and activator of transcription 3 (STAT3) degradation by proteasome controls a developmental switch in neurotrophin dependence. J Biol Chem 2013; 288:20151-61. [PMID: 23733189 DOI: 10.1074/jbc.m113.470583] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neonatal brains develop through a program that eliminates about half of the neurons. During this period, neurons depend on neurotrophins for their survival. Recently, we reported that, at the conclusion of the naturally occurring death period, neurons become neurotrophin-independent and, further, that this developmental switch is achieved by the emergence of a second survival pathway mediated by signal transducer and activator of transcription 3 (STAT3). Here I show that calcineurin plays a key role in controlling the developmental switch in mouse hippocampal neurons. Calcineurin promotes the degradation of STAT3 via the ubiquitin-proteasome pathway. Inhibition of calcineurin acutely increases total levels of STAT3 as well as its activated forms, resulting in decreased levels of the tumor suppressor p53 and its proapoptotic target, Bax. In vivo and in vitro, calcineurin regulates levels of STAT3 and neurotrophin dependence. TMF/ARA 160 (TATA element modulatory factor/androgen receptor co-activator 160), the key mediator of STAT3 ubiquitination, is required for calcineurin-dependent STAT3 degradation. Thus, these results show that the ubiquitin-proteasome pathway controls the critical developmental switch of neurotrophin dependence in the newborn hippocampus.
Collapse
Affiliation(s)
- Sachiko Murase
- Laboratory of Molecular Biology, NINDS, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|
45
|
Rojanathammanee L, Puig KL, Combs CK. Pomegranate polyphenols and extract inhibit nuclear factor of activated T-cell activity and microglial activation in vitro and in a transgenic mouse model of Alzheimer disease. J Nutr 2013; 143:597-605. [PMID: 23468550 PMCID: PMC3738232 DOI: 10.3945/jn.112.169516] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Alzheimer disease (AD) brain is characterized by extracellular plaques of amyloid β (Aβ) peptide with reactive microglia. This study aimed to determine whether a dietary intervention could attenuate microgliosis. Memory was assessed in 12-mo-old male amyloid precursor protein/presenilin 1 (APP/PS1) transgenic mice via Barnes maze testing followed by division into either a control-fed group provided free access to normal chow and water or a treatment group provided free access to normal chow and drinking water supplemented with pomegranate extract (6.25 mL/L) for 3 mo followed by repeat Barnes maze testing for both groups. Three months of pomegranate feeding decreased the path length to escape of mice compared with their initial 12-mo values (P < 0.05) and their control-fed counterparts (P < 0.05). Brains of the 3-mo study pomegranate-fed mice had lower tumor necrosis factor α (TNF-α) concentrations (P < 0.05) and lower nuclear factor of activated T-cell (NFAT) transcriptional activity (P < 0.05) compared with controls. Brains of the 3-mo pomegranate or control mice were also compared with an additional control group of 12-mo-old mice for histologic analysis. Immunocytochemistry showed that pomegranate- but not control-fed mice had attenuated microgliosis (P < 0.05) and Aβ plaque deposition (P < 0.05) compared with 12-mo-old mice. An additional behavioral study again used 12-mo-old male APP/PS1 mice tested by T-maze followed by division into a control group provided with free access to normal chow and sugar supplemented drinking water or a treatment group provided with normal chow and pomegranate extract-supplemented drinking water (6.25 mL/L) for 1 mo followed by repeat T-maze testing in both groups. One month of pomegranate feeding increased spontaneous alternations versus control-fed mice (P < 0.05). Cell culture experiments verified that 2 polyphenol components of pomegranate extract, punicalagin and ellagic acid, attenuated NFAT activity in a reporter cell line (P < 0.05) and decreased Aβ-stimulated TNF-α secretion by murine microglia (P < 0.05). These data indicate that dietary pomegranate produces brain antiinflammatory effects that may attenuate AD progression.
Collapse
|
46
|
Temporal regulation of nuclear factor one occupancy by calcineurin/NFAT governs a voltage-sensitive developmental switch in late maturing neurons. J Neurosci 2013; 33:2860-72. [PMID: 23407945 DOI: 10.1523/jneurosci.3533-12.2013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Dendrite and synapse development are critical for establishing appropriate neuronal circuits, and disrupted timing of these events can alter neural connectivity. Using microarrays, we have identified a nuclear factor I (NFI)-regulated temporal switch program linked to dendrite formation in developing mouse cerebellar granule neurons (CGNs). NFI function was required for upregulation of many synapse-related genes as well as downregulation of genes expressed in immature CGNs. Chromatin immunoprecipitation analysis revealed that a central feature of this program was temporally regulated NFI occupancy of late-expressed gene promoters. Developing CGNs undergo a hyperpolarizing shift in membrane potential, and depolarization inhibits their dendritic and synaptic maturation via activation of calcineurin (CaN) (Okazawa et al., 2009). Maintaining immature CGNs in a depolarized state blocked NFI temporal occupancy of late-expressed genes and the NFI switch program via activation of the CaN/nuclear factor of activated T-cells, cytoplasmic (NFATc) pathway and promotion of late-gene occupancy by NFATc4, and these mechanisms inhibited dendritogenesis. Conversely, inhibition of the CaN/NFATc pathway in CGNs maturing under physiological nondepolarizing conditions upregulated the NFI switch program, NFI temporal occupancy, and dendrite formation. NFATc4 occupied the promoters of late-expressed NFI program genes in immature mouse cerebellum, and its binding was temporally downregulated with development. Further, NFI temporal binding and switch gene expression were upregulated in the developing cerebellum of Nfatc4 (-/-) mice. These findings define a novel NFI switch and temporal occupancy program that forms a critical link between membrane potential/CaN and dendritic maturation in CGNs. CaN inhibits the program and NFI occupancy in immature CGNs by promoting NFATc4 binding to late-expressed genes. As maturing CGNs become more hyperpolarized, NFATc4 binding declines leading to onset of NFI temporal binding and the NFI switch program.
Collapse
|
47
|
Li R, Zhang L, Shi W, Zhang B, Liang X, Liu S, Wang W. NFAT2 mediates high glucose-induced glomerular podocyte apoptosis through increased Bax expression. Exp Cell Res 2013; 319:992-1000. [PMID: 23340267 DOI: 10.1016/j.yexcr.2013.01.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Revised: 12/25/2012] [Accepted: 01/07/2013] [Indexed: 01/19/2023]
Abstract
BACKGROUND Hyperglycemia promotes podocyte apoptosis and plays a key role in the pathogenesis of diabetic nephropathy. However, the mechanisms that mediate hyperglycemia-induced podocyte apoptosis is still far from being fully understood. Recent studies reported that high glucose activate nuclear factor of activated T cells (NFAT) in vascular smooth muscle or pancreatic β-cells. Here, we sought to determine if hyperglycemia activates NFAT2 in cultured podocyte and whether this leads to podocyte apoptosis. Meanwhile, we also further explore the mechanisms of NFAT2 activation and NFAT2 mediates high glucose-induced podocyte apoptosis. METHODS Immortalized mouse podocytes were cultured in media containing normal glucose (NG), or high glucose (HG) or HG plus cyclosporine A (a pharmacological inhibitor of calcinerin) or 11R-VIVIT (a special inhibitor of NFAT2). The activation of NFAT2 in podocytes was detected by western blotting and immunofluorescence assay. The role of NFAT2 in hyperglycemia-induced podocyte apoptosis was further evaluated by observing the inhibition of NFAT2 activation by 11R-VIVIT using flow cytometer. Intracellular Ca(2+) was monitored in HG-treated podcocytes using Fluo-3/AM. The mRNA and protein expression of apoptosis gene Bax were measured by real time-qPCR and western blotting. RESULTS HG stimulation activated NFAT2 in a time- and dose-dependent manner in cultured podocytes. Pretreatment with cyclosporine A (500nM) or 11R-VIVIT (100nM) completely blocked NFAT2 nuclear accumulation. Meanwhile, the apoptosis effects induced by HG were also abrogated by concomitant treatment with 11R-VIVIT in cultured podocytes. We further found that HG also increased [Ca(2+)]i, leading to activation of calcineurin, and subsequent increased nuclear accumulation of NFAT2 and Bax expression in cultured podocytes. CONCLUSION Our results identify a new finding that HG-induced podocyte apoptosis is mediated by calcineurin/NFAT2/Bax signaling pathway, which may present a promising target for therapeutic intervention.
Collapse
Affiliation(s)
- Ruizhao Li
- Department of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan No. 2 Road, Guangzhou 510080, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
48
|
Ambacher KK, Pitzul KB, Karajgikar M, Hamilton A, Ferguson SS, Cregan SP. The JNK- and AKT/GSK3β- signaling pathways converge to regulate Puma induction and neuronal apoptosis induced by trophic factor deprivation. PLoS One 2012; 7:e46885. [PMID: 23056511 PMCID: PMC3463558 DOI: 10.1371/journal.pone.0046885] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 09/07/2012] [Indexed: 12/17/2022] Open
Abstract
The AKT, GSK3 and JNK family kinases have been implicated in neuronal apoptosis associated with neuronal development and several neurodegenerative conditions. However, the mechanisms by which these kinase pathways regulate apoptosis remain unclear. In this study we have investigated the role of these kinases in neuronal cell death using an established model of trophic factor deprivation induced apoptosis in cerebellar granule neurons. BCL-2 family proteins are known to be central regulators of apoptosis and we have determined that the pro-apoptotic family member Puma is transcriptionally up-regulated in trophic factor deprived neurons and that Puma induction is required for apoptosis in vitro and in vivo. Importantly, we demonstrate that Puma induction is dependent on both JNK activation and AKT inactivation. AKT is known to regulate a number of downstream pathways, however we have determined that PI3K-AKT inactivation induces Puma expression through a GSK3β-dependent mechanism. Finally we demonstrate that the JNK and AKT/GSK3β pathways converge to regulate FoxO3a-mediated transcriptional activation of Puma. In summary we have identified a novel and critical link between the AKT, GSK3β and JNK kinases and the regulation of Puma induction and suggest that this may be pivotal to the regulation of neuronal apoptosis in neurodegenerative conditions.
Collapse
Affiliation(s)
- Kristin K. Ambacher
- Robarts Research Institute and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Kristen B. Pitzul
- Robarts Research Institute and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Meera Karajgikar
- Robarts Research Institute and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Alison Hamilton
- Robarts Research Institute and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Stephen S. Ferguson
- Robarts Research Institute and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Sean P. Cregan
- Robarts Research Institute and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
49
|
Ulrich JD, Kim MS, Houlihan PR, Shutov LP, Mohapatra DP, Strack S, Usachev YM. Distinct activation properties of the nuclear factor of activated T-cells (NFAT) isoforms NFATc3 and NFATc4 in neurons. J Biol Chem 2012; 287:37594-609. [PMID: 22977251 DOI: 10.1074/jbc.m112.365197] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The Ca(2+)/calcineurin-dependent transcription factor NFAT (nuclear factor of activated T-cells) is implicated in regulating dendritic and axonal development, synaptogenesis, and neuronal survival. Despite the increasing appreciation for the importance of NFAT-dependent transcription in the nervous system, the regulation and function of specific NFAT isoforms in neurons are poorly understood. Here, we compare the activation of NFATc3 and NFATc4 in hippocampal and dorsal root ganglion neurons following electrically evoked elevations of intracellular Ca(2+) concentration ([Ca(2+)](i)). We find that NFATc3 undergoes rapid dephosphorylation and nuclear translocation that are essentially complete within 20 min, although NFATc4 remains phosphorylated and localized to the cytosol, only exhibiting nuclear localization following prolonged (1-3 h) depolarization. Knocking down NFATc3, but not NFATc4, strongly diminished NFAT-mediated transcription induced by mild depolarization in neurons. By analyzing NFATc3/NFATc4 chimeras, we find that the region containing the serine-rich region-1 (SRR1) mildly affects initial NFAT translocation, although the region containing the serine-proline repeats is critical for determining the magnitude of NFAT activation and nuclear localization upon depolarization. Knockdown of glycogen synthase kinase 3β (GSK3β) significantly increased the depolarization-induced nuclear localization of NFATc4. In contrast, inhibition of p38 or mammalian target of rapamycin (mTOR) kinases had no significant effect on nuclear import of NFATc4. Thus, electrically evoked [Ca(2+)](i) elevation in neurons rapidly and strongly activates NFATc3, whereas activation of NFATc4 requires a coincident increase in [Ca(2+)](i) and suppression of GSK3β, with differences in the serine-proline-containing region giving rise to these distinct activation properties of NFATc3 and NFATc4.
Collapse
Affiliation(s)
- Jason D Ulrich
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Inhibition of the NFAT pathway alleviates amyloid β neurotoxicity in a mouse model of Alzheimer's disease. J Neurosci 2012; 32:3176-92. [PMID: 22378890 DOI: 10.1523/jneurosci.6439-11.2012] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Amyloid β (Aβ) peptides, the main pathological species associated with Alzheimer's disease (AD), disturb intracellular calcium homeostasis, which in turn activates the calcium-dependent phosphatase calcineurin (CaN). CaN activation induced by Aβ leads to pathological morphological changes in neurons, and overexpression of constitutively active calcineurin is sufficient to generate a similar phenotype, even without Aβ. Here, we tested the hypothesis that calcineurin mediates neurodegenerative effects via activation of the nuclear transcription factor of activated T-cells (NFAT). We found that both spine loss and dendritic branching simplification induced by Aβ exposure were mimicked by constitutively active NFAT, and abolished when NFAT activation was blocked using the genetically encoded inhibitor VIVIT. When VIVIT was specifically addressed to the nucleus, identical beneficial effects were observed, thus enforcing the role of NFAT transcriptional activity in Aβ-related neurotoxicity. In vivo, when VIVIT or its nuclear counterpart were overexpressed in a transgenic model of Alzheimer's disease via a gene therapy approach, the spine loss and neuritic abnormalities observed in the vicinity of amyloid plaques were blocked. Overall, these results suggest that NFAT/calcineurin transcriptional cascades contribute to Aβ synaptotoxicity, and may provide a new specific set of pathways for neuroprotective strategies.
Collapse
|