1
|
He X, Sun X, Shao Y. Multicellular Network-Informed Survival Model for Identification of Drug Targets of Gliomas. IEEE J Biomed Health Inform 2025; 29:1591-1601. [PMID: 37643106 DOI: 10.1109/jbhi.2023.3309825] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Increasing evidence suggests that communication between tumor cells (TCs) and tumor-associated macrophages (TAMs) plays a substantial role in promoting progression of low-grade gliomas (LGG). Hence, it is becoming critical to model TAM-TC interplay and interrogate how the crosstalk affects prognosis of LGG patients. This article proposed a translational research pipeline to construct the multicellular interaction gene network (MIGN) for identification of druggable targets to develop novel therapeutic strategies. Firstly, we selected immunotherapy-related feature genes (IFGs) for TAMs and TCs using RNA-seq data of glioma mice from preclinical trials. After translating the IFGs to human genome, we constructed TAM- and TC- associated networks separately, using a training set of 524 human LGGs. Subsequently, clustering analysis was performed within each network, and the concordance measure K-index was adopted to correlate gene clusters with patient survival. The MIGN was built by combining the clusters highly associated with survival in TAM- and TC-associated networks. We then developed a MIGN-based survival model to identify prognostic signatures comprised of ligands, receptors and hub genes. An independent cohort of 172 human LGG samples was leveraged to validate predictive accuracy of the signature. The areas under time-dependent ROC curves were 0.881, 0.867, and 0.839 with respect to 1-year, 3-year, and 5-year survival rates respectively in the validation set. Furthermore, literature survey was conducted on the signature genes, and potential clinical responses to targeted drugs were evaluated for LGG patients, further highlighting potential utilities of the MIGN signature to develop novel immunotherapies to extend survival of LGG patients.
Collapse
|
2
|
Shankar S, Liu Y, Tulsian NK, Low BC, Lin Q, Sivaraman J. Insights into the regulation of CHIP E3 ligase-mediated ubiquitination of neuronal protein BNIP-H. PNAS NEXUS 2024; 3:pgae536. [PMID: 39703232 PMCID: PMC11658413 DOI: 10.1093/pnasnexus/pgae536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/04/2024] [Indexed: 12/21/2024]
Abstract
BCL2/adenovirus E1B 19-kDa protein-interacting protein 2 homolog (BNIP-H or Caytaxin), a pivotal adaptor protein that facilitates cerebellar cortex growth and synaptic transmission, is posttranslationally modified to regulate neuronal function. This study reports the ubiquitination of BNIP-H by Carboxyl terminus of Hsc70-Interacting Protein (CHIP), a U-box containing E3 ligase that is also regulated via autoubiquitination. Specifically, it was observed that CHIP autoubiquitinated itself primarily at Lys23 and Lys31 in vitro. Mutation of these residues shows the autoubiquitination of successive lysines of CHIP. In total, nine lysines on CHIP were identified as the autoubiquitination sites, the collective mutation of which almost completely terminated its autoubiquitination. Additionally, CHIP-mediated ubiquitination of BNIP-H is completely inhibited when BNIP-H bears arginine mutations at four key lysine residues. Next, using hydrogen deuterium exchange mass spectrometry, a model of a plausible mechanism was proposed. The model suggests transient N-terminal interactions between the CHIP and BNIP-H which allows for the swinging of U-box domain of CHIP to ubiquitinate BNIP-H. Following complex dissociation, BNIP-H population is regulated via the ubiquitin-proteasome pathway. Collectively, these results aid in our understanding of CHIP-mediated BNIP-H ubiquitination and provide further insight into the roles of these proteins in neuritogenesis and neurotransmission.
Collapse
Affiliation(s)
- Srihari Shankar
- Department of Biological Sciences, National University of Singapore, Singapore 117543
| | - Yaochen Liu
- Department of Biological Sciences, National University of Singapore, Singapore 117543
| | - Nikhil Kumar Tulsian
- Department of Biological Sciences, National University of Singapore, Singapore 117543
| | - Boon C Low
- Department of Biological Sciences, National University of Singapore, Singapore 117543
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- NUS College, National University of Singapore, Singapore 138593
| | - Qingsong Lin
- Department of Biological Sciences, National University of Singapore, Singapore 117543
| | - J Sivaraman
- Department of Biological Sciences, National University of Singapore, Singapore 117543
| |
Collapse
|
3
|
Shankar S, Chew TW, Chichili VPR, Low BC, Sivaraman J. Structural basis for the distinct roles of non-conserved Pro116 and conserved Tyr124 of BCH domain of yeast p50RhoGAP. Cell Mol Life Sci 2024; 81:216. [PMID: 38740643 PMCID: PMC11090974 DOI: 10.1007/s00018-024-05238-8] [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: 10/25/2023] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 05/16/2024]
Abstract
p50RhoGAP is a key protein that interacts with and downregulates the small GTPase RhoA. p50RhoGAP is a multifunctional protein containing the BNIP-2 and Cdc42GAP Homology (BCH) domain that facilitates protein-protein interactions and lipid binding and the GAP domain that regulates active RhoA population. We recently solved the structure of the BCH domain from yeast p50RhoGAP (YBCH) and showed that it maintains the adjacent GAP domain in an auto-inhibited state through the β5 strand. Our previous WT YBCH structure shows that a unique kink at position 116 thought to be made by a proline residue between alpha helices α6 and α7 is essential for the formation of intertwined dimer from asymmetric monomers. Here we sought to establish the role and impact of this Pro116. However, the kink persists in the structure of P116A mutant YBCH domain, suggesting that the scaffold is not dictated by the proline residue at this position. We further identified Tyr124 (or Tyr188 in HBCH) as a conserved residue in the crucial β5 strand. Extending to the human ortholog, when substituted to acidic residues, Tyr188D or Tyr188E, we observed an increase in RhoA binding and self-dimerization, indicative of a loss of inhibition of the GAP domain by the BCH domain. These results point to distinct roles and impact of the non-conserved and conserved amino acid positions in regulating the structural and functional complexity of the BCH domain.
Collapse
Affiliation(s)
- Srihari Shankar
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore
| | - Ti Weng Chew
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
| | | | - Boon Chuan Low
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore.
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore.
- NUS College, National University of Singapore, Singapore, 138593, Singapore.
| | - J Sivaraman
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore.
| |
Collapse
|
4
|
Siavashani ES, Ashrafi MR, Ghabeli H, Heidari M, Garshasbi M. Novel homozygous frameshift variant in the ATCAY gene in an Iranian patient with Cayman cerebellar ataxia; expanding the neuroimaging and clinical features: a case report. BMC Med Genomics 2023; 16:226. [PMID: 37752557 PMCID: PMC10523697 DOI: 10.1186/s12920-023-01643-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Pathogenic variants in the ATCAY gene are associated with a rare autosomal recessive disorder called Cayman cerebellar ataxia. Affected individuals display psychomotor retardation, cerebellar dysfunction, nystagmus, intention tremor, ataxic gait and dysarthric in some cases. CASE PRESENTATION Whole exome sequencing was performed for a 21-month-old girl suffering from developmental delay specifically in motor and language aspects, hypotonia, nystagmus, pes planus and strabismus. The detected variant in the patient was confirmed by family segregation analysis by Sanger sequencing in both of her parents. Previously three homozygous variants in the ATCAY gene (missense, splice site and frameshift deletion) have been reported in patients with Cayman cerebellar ataxia. Here we report the fourth homozygous variant and the second homozygous frameshift deletion in this gene to be associated with autosomal recessive Cayman cerebellar ataxia. CONCLUSION The identification of this novel homozygous frameshift deletion in the ATCAY gene expands our understanding of the genetic landscape underlying Cayman cerebellar ataxia. Furthermore, the occurrence of this variant in Iran, in addition to Pakistan, signifies the importance of considering genotypic and phenotypic factors beyond ethnicity when studying this disorder. These findings contribute to the ongoing efforts to unravel the molecular basis of Cayman cerebellar ataxia and improve diagnostic approaches and potential therapeutic interventions.
Collapse
Affiliation(s)
- Elham Salehi Siavashani
- PardisGene Co., Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahmoud Reza Ashrafi
- Department of Pediatric Neurology, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Homa Ghabeli
- Pediatric Neurology Division, Myelin Disorders Clinic, Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Heidari
- Department of Pediatric Neurology, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Pediatric Neurology Division, Myelin Disorders Clinic, Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran.
| | - Masoud Garshasbi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
5
|
Pereira CPM, Francis-Oliveira J, Singulani MP, Ferreira AFF, Britto LRG. Microglial depletion exacerbates motor impairment and dopaminergic neuron loss in a 6-OHDA model of Parkinson's disease. J Neuroimmunol 2023; 375:578019. [PMID: 36681049 DOI: 10.1016/j.jneuroim.2023.578019] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/28/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
6-hydroxydopamine (6-OHDA) is a common neurotoxin used to induce Parkinson's disease (PD) in mice, exerting neurotoxic effects through the production of reactive oxygen species and microglial activation. However, the role of microglia in PD is still not clear, with contradictory reports showing neuroprotection or exacerbation of neuronal death. Microglial depletion aggravates motor coordination impairments and reduces tyrosine hydroxylase positive neurons in the substantia nigra pars compacta. Moreover, MeCP2 and Adora1 genes expression were downregulated, suggesting they may be involved in the neurodegenerative process. This study highlights that microglia plays a protective role in dopaminergic neuron survival during the initial phase of PD, and the investigation of the mechanisms of this effect in future studies will help elucidate the pathophysiology of PD.
Collapse
Affiliation(s)
- Carolina Parga Martins Pereira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil; Department of Neurobiology and Behavior, Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, USA.
| | - José Francis-Oliveira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil; Departament of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, USA
| | - Monique Patricio Singulani
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Ana Flávia Fernandes Ferreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Roberto G Britto
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
6
|
Shim SM, Koh YH, Kim JH, Jeon JP. A combination of multiple autoantibodies is associated with the risk of Alzheimer’s disease and cognitive impairment. Sci Rep 2022; 12:1312. [PMID: 35079008 PMCID: PMC8789802 DOI: 10.1038/s41598-021-04556-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 12/06/2021] [Indexed: 11/02/2022] Open
Abstract
AbstractAutoantibodies are self-antigen reactive antibodies that play diverse roles in the normal immune system, tissue homeostasis, and autoimmune and neurodegenerative diseases. Anti-neuronal autoantibodies have been detected in neurodegenerative disease serum, with unclear significance. To identify diagnostic biomarkers of Alzheimer’s disease (AD), we analyzed serum autoantibody profiles of the HuProt proteome microarray using the discovery set of cognitively normal control (NC, n = 5) and AD (n = 5) subjects. Approximately 1.5-fold higher numbers of autoantibodies were detected in the AD group (98.0 ± 39.9/person) than the NC group (66.0 ± 39.6/person). Of the autoantigen candidates detected in the HuProt microarray, five autoantigens were finally selected for the ELISA-based validation experiment using the validation set including age- and gender-matched normal (NC, n = 44), mild cognitive impairment (MCI, n = 44) and AD (n = 44) subjects. The serum levels of four autoantibodies including anti-ATCAY, HIST1H3F, NME7 and PAIP2 IgG were significantly different among NC, MCI and/or AD groups. Specifically, the anti-ATCAY autoantibody level was significantly higher in the AD (p = 0.003) and MCI (p = 0.015) groups compared to the NC group. The anti-ATCAY autoantibody level was also significantly correlated with neuropsychological scores of MMSE (rs = − 0.229, p = 0.012), K-MoCA (rs = − 0.270, p = 0.003), and CDR scores (rs = 0.218, p = 0.016). In addition, a single or combined occurrence frequency of anti-ATCAY and anti-PAIP2 autoantibodies was significantly associated with the risk of MCI and AD. This study indicates that anti-ATCAY and anti-PAIP2 autoantibodies could be a potential diagnostic biomarker of AD.
Collapse
|
7
|
Structural basis for p50RhoGAP BCH domain-mediated regulation of Rho inactivation. Proc Natl Acad Sci U S A 2021; 118:2014242118. [PMID: 34006635 DOI: 10.1073/pnas.2014242118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Spatiotemporal regulation of signaling cascades is crucial for various biological pathways, under the control of a range of scaffolding proteins. The BNIP-2 and Cdc42GAP Homology (BCH) domain is a highly conserved module that targets small GTPases and their regulators. Proteins bearing BCH domains are key for driving cell elongation, retraction, membrane protrusion, and other aspects of active morphogenesis during cell migration, myoblast differentiation, and neuritogenesis. We previously showed that the BCH domain of p50RhoGAP (ARHGAP1) sequesters RhoA from inactivation by its adjacent GAP domain; however, the underlying molecular mechanism for RhoA inactivation by p50RhoGAP remains unknown. Here, we report the crystal structure of the BCH domain of p50RhoGAP Schizosaccharomyces pombe and model the human p50RhoGAP BCH domain to understand its regulatory function using in vitro and cell line studies. We show that the BCH domain adopts an intertwined dimeric structure with asymmetric monomers and harbors a unique RhoA-binding loop and a lipid-binding pocket that anchors prenylated RhoA. Interestingly, the β5-strand of the BCH domain is involved in an intermolecular β-sheet, which is crucial for inhibition of the adjacent GAP domain. A destabilizing mutation in the β5-strand triggers the release of the GAP domain from autoinhibition. This renders p50RhoGAP active, thereby leading to RhoA inactivation and increased self-association of p50RhoGAP molecules via their BCH domains. Our results offer key insight into the concerted spatiotemporal regulation of Rho activity by BCH domain-containing proteins.
Collapse
|
8
|
Wang S, Chen K, Yu J, Wang X, Li Q, Lv F, Shen H, Pei L. Presynaptic Caytaxin prevents apoptosis via deactivating DAPK1 in the acute phase of cerebral ischemic stroke. Exp Neurol 2020; 329:113303. [PMID: 32277960 DOI: 10.1016/j.expneurol.2020.113303] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 03/19/2020] [Accepted: 04/07/2020] [Indexed: 12/19/2022]
Abstract
Death-associated protein kinase 1 (DAPK1) is a key protein that mediates neuronal death in ischemic stroke. Although the substrates of DAPK1 and molecular signal in stroke have been gradually discovered, the modulation of DAPK1 itself is still unclear. Here we first reveal that Caytaxin, a brain-specific member of BCL2/adenovirus E1B -interacting protein (BNIP-2), increases and interacts with DAPK1 as early as 2 h after middle cerebral artery occlusion (MCAO) in the penumbra area of mouse brain. Furthermore, Caytaxin binds to DAPK1 at the presynaptic site and inhibits DAPK1 catalytic activity. Silencing Caytaxin by Caytaxin shRNA (Sh-Caytaxin) enhances DAPK1 activity, deteriorates neuronal apoptosis and brain injuries both in vivo and in vitro. Thus, elevating presynaptic Caytaxin could prevent neuronal apoptosis by inhibiting DAPK1 activation in the acute stage of ischemic stroke. Caytaxin may physiologically protect neuronal cells and represent a potential prevention and therapeutic target in the early phase of cerebral ischemic stroke.
Collapse
Affiliation(s)
- Shan Wang
- Department of Biotherapy Center, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shen Zhen 518033, China
| | - Keng Chen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, China
| | - Jia Yu
- Exchange, Development & Service Center for Science & Technology Talents, The Ministry of Science and Technology (Most), Beijing 100045, China
| | - Xiaojiao Wang
- Exchange, Development & Service Center for Science & Technology Talents, The Ministry of Science and Technology (Most), Beijing 100045, China
| | - Qiang Li
- Exchange, Development & Service Center for Science & Technology Talents, The Ministry of Science and Technology (Most), Beijing 100045, China
| | - Fei Lv
- Department of Emergency, Wuhan Asia Heart Hospital, Wuhan 430033, China
| | - Huiyong Shen
- Department of Biotherapy Center, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shen Zhen 518033, China; Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, China.
| | - Lei Pei
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
9
|
Hoerner CR, Chen VJ, Fan AC. The 'Achilles Heel' of Metabolism in Renal Cell Carcinoma: Glutaminase Inhibition as a Rational Treatment Strategy. KIDNEY CANCER 2019; 3:15-29. [PMID: 30854496 PMCID: PMC6400133 DOI: 10.3233/kca-180043] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
An important hallmark of cancer is 'metabolic reprogramming' or the rewiring of cellular metabolism to support rapid cell proliferation [1-5]. Metabolic reprogramming through oncometabolite-mediated transformation or activation of oncogenes in renal cell carcinoma (RCC) globally impacts energy production as well as glucose and glutamine utilization in RCC cells, which can promote dependence on glutamine supply to support cell growth and proliferation [6, 7]. Novel inhibitors of glutaminase, a key enzyme in glutamine metabolism, target glutamine addiction as a viable treatment strategy in metastatic RCC (mRCC). Here, we review glutamine metabolic pathways and how changes in cellular glutamine utilization enable the progression of RCC. This overview provides scientific rationale for targeting this pathway in patients with mRCC. We will summarize the current understanding of cellular and molecular mechanisms underlying anti-tumor efficacy of glutaminase inhibitors in RCC, provide an overview of clinical efforts targeting glutaminase in mRCC, and review approaches for identifying biomarkers for patient stratification and detecting therapeutic response early on in patients treated with this novel class of anti-cancer drug. Ultimately, results of ongoing clinical trials will demonstrate whether glutaminase inhibition can be a worthy addition to the current armamentarium of drugs used for patients with mRCC.
Collapse
Affiliation(s)
- Christian R Hoerner
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, CA, USA
| | - Viola J Chen
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, CA, USA
| | - Alice C Fan
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, CA, USA
| |
Collapse
|
10
|
de Guzzi Cassago CA, Dias MM, Pinheiro MP, Pasquali CC, Bastos ACS, Islam Z, Consonni SR, de Oliveira JF, Gomes EM, Ascenção CFR, Honorato R, Pauletti BA, Indolfo NDC, Filho HVR, de Oliveira PSL, Figueira ACM, Paes Leme AF, Ambrosio ALB, Dias SMG. Glutaminase Affects the Transcriptional Activity of Peroxisome Proliferator-Activated Receptor γ (PPARγ) via Direct Interaction. Biochemistry 2018; 57:6293-6307. [PMID: 30295466 DOI: 10.1021/acs.biochem.8b00773] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Phosphate-activated glutaminases catalyze the deamidation of glutamine to glutamate and play key roles in several physiological and pathological processes. In humans, GLS encodes two multidomain splicing isoforms: KGA and GAC. In both isoforms, the canonical glutaminase domain is flanked by an N-terminal region that is folded into an EF-hand-like four-helix bundle. However, the splicing event replaces a well-structured three-repeat ankyrin domain in KGA with a shorter, unordered C-terminal stretch in GAC. The multidomain architecture, which contains putative protein-protein binding motifs, has led to speculation that glutaminases are involved in cellular processes other than glutamine metabolism; in fact, some proteins have been identified as binding partners of KGA and the isoforms of its paralogue gene, GLS2. Here, a yeast two-hybrid assay identified nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) as a new binding partner of the glutaminase. We show that KGA and GAC directly bind PPARγ with a low-micromolar dissociation constant; the interaction involves the N-terminal and catalytic domains of glutaminases as well as the ligand-binding domain of the nuclear receptor. The interaction occurs within the nucleus, and by sequestering PPARγ from its responsive element DR1, the glutaminases decreased nuclear receptor activity as assessed by a luciferase reporter assay. Altogether, our findings reveal an unexpected glutaminase-binding partner and, for the first time, directly link mitochondrial glutaminases to an unanticipated role in gene regulation.
Collapse
Affiliation(s)
- Carolina Aparecida de Guzzi Cassago
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Marília Meira Dias
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Matheus Pinto Pinheiro
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Camila Cristina Pasquali
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Alliny Cristiny Silva Bastos
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology , University of Campinas (UNICAMP) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Zeyaul Islam
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Sílvio Roberto Consonni
- Department of Biochemistry and Tissue Biology , University of Campinas , Campinas , Sao Paulo 13083-872 , Brazil
| | - Juliana Ferreira de Oliveira
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Emerson Machi Gomes
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Carolline Fernanda Rodrigues Ascenção
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology , University of Campinas (UNICAMP) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Rodrigo Honorato
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Bianca Alves Pauletti
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Nathalia de Carvalho Indolfo
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Helder Veras Ribeiro Filho
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Paulo Sergio Lopes de Oliveira
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Ana Carolina Migliorini Figueira
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Adriana Franco Paes Leme
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Andre Luis Berteli Ambrosio
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Sandra Martha Gomes Dias
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| |
Collapse
|
11
|
Guo Y, Li H, Ke X, Deng M, Wu Z, Cai Y, Afewerky HK, Zhang X, Pei L, Lu Y. Degradation of Caytaxin Causes Learning and Memory Deficits via Activation of DAPK1 in Aging. Mol Neurobiol 2018; 56:3368-3379. [PMID: 30120735 DOI: 10.1007/s12035-018-1312-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/08/2018] [Indexed: 12/11/2022]
Abstract
Loss of memory is an inevitable clinic sign in aging, but its underlying mechanisms remain unclear. Here we show that death-associated protein kinase (DAPK1) is involved in the decays of learning and memory in aging via degradation of Caytaxin, a brain-specific member of BNIP-2. DAPK1 becomes activated in the hippocampus of mice during aging. Activation of DAPK1 is closely associated with degradation of Caytaxin protein. Silencing Caytaxin by the expression of small interfering RNA (siRNA) that targets specifically to Caytaxin in the hippocampus of adult mice impairs the learning and memory. Genetic inactivation of DAPK1 by deletion of DAPK1 kinase domain prevents the degradation of Caytaxin and protects against learning and memory declines. Thus, activation of DAPK1 impairs learning and memory by degrading Caytaxin during aging.
Collapse
Affiliation(s)
- Yu Guo
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hao Li
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Ke
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Manfei Deng
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhuoze Wu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - You Cai
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Henok Kessete Afewerky
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Pathology and Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaoan Zhang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lei Pei
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.
| | - Youming Lu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China. .,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
12
|
Islam S, Ueda M, Nishida E, Wang MX, Osawa M, Lee D, Itoh M, Nakagawa K, Tana, Nakagawa T. Odor preference and olfactory memory are impaired in Olfaxin-deficient mice. Brain Res 2018; 1688:81-90. [PMID: 29571668 DOI: 10.1016/j.brainres.2018.03.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/19/2018] [Accepted: 03/19/2018] [Indexed: 12/12/2022]
Abstract
Olfaxin, which is a BNIP2 and Cdc42GAP homology (BCH) domain-containing protein, is predominantly expressed in mitral and tufted (M/T) cells in the olfactory bulb (OB). Olfaxin and Caytaxin, which share 56.3% amino acid identity, are similar in their glutamatergic terminal localization, kidney-type glutaminase (KGA) interaction, and caspase-3 substrate. Although the deletion of Caytaxin protein causes human Cayman ataxia and ataxia in the mutant mouse, the function of Olfaxin is largely unknown. In this study, we generated Prune2 gene mutant mice (Prune2Ex16-/-; knock out [KO] mice) using the CRISPR/Cas9 system, during which the exon 16 containing start codon of Olfaxin mRNA was deleted. Exon 16 has 80 nucleotides and is contained in four of five Prune2 isoforms, including PRUNE2, BMCC1, BNIPXL, and Olfaxin/BMCC1s. The levels of Olfaxin mRNA and Olfaxin protein in the OB and piriform cortex of KO mice significantly decreased. Although Prune2 mRNA also significantly decreased in the spinal cord, the gross anatomy of the spinal cord and dorsal root ganglion (DRG) was intact. Further, disturbance of the sensory and motor system was not observed in KO mice. Therefore, in the current study, we examined the role of Olfaxin in the olfactory system where PRUNE2, BMCC1, and BNIPXL are scarcely expressed. Odor preference was impaired in KO mice using opposite-sex urinary scents as well as a non-social odor stimulus (almond). Results of the odor-aversion test demonstrated that odor-associative learning was disrupted in KO mice. Moreover, the NMDAR2A/NMDAR2B subunits switch in the piriform cortex was not observed in KO mice. These results indicated that Olfaxin may play a critical role in odor preference and olfactory memory.
Collapse
Affiliation(s)
- Saiful Islam
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masashi Ueda
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan; Department of Embryology, Institute for Developmental Research, Aichi Human Service Center, Aichi, Japan
| | - Emika Nishida
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Miao-Xing Wang
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masatake Osawa
- Department of Molecular Design and Synthesis, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Dongsoo Lee
- Department of Molecular Design and Synthesis, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masanori Itoh
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kiyomi Nakagawa
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tana
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Toshiyuki Nakagawa
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, Japan.
| |
Collapse
|
13
|
Peñalver A, Campos-Sandoval JA, Blanco E, Cardona C, Castilla L, Martín-Rufián M, Estivill-Torrús G, Sánchez-Varo R, Alonso FJ, Pérez-Hernández M, Colado MI, Gutiérrez A, de Fonseca FR, Márquez J. Glutaminase and MMP-9 Downregulation in Cortex and Hippocampus of LPA 1 Receptor Null Mice Correlate with Altered Dendritic Spine Plasticity. Front Mol Neurosci 2017; 10:278. [PMID: 28928633 PMCID: PMC5591874 DOI: 10.3389/fnmol.2017.00278] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/17/2017] [Indexed: 12/03/2022] Open
Abstract
Lysophosphatidic acid (LPA) is an extracellular lipid mediator that regulates nervous system development and functions acting through G protein-coupled receptors (GPCRs). Here we explore the crosstalk between LPA1 receptor and glutamatergic transmission by examining expression of glutaminase (GA) isoforms in different brain areas isolated from wild-type (WT) and KOLPA1 mice. Silencing of LPA1 receptor induced a severe down-regulation of Gls-encoded long glutaminase protein variant (KGA) (glutaminase gene encoding the kidney-type isoforms, GLS) protein expression in several brain regions, particularly in brain cortex and hippocampus. Immunohistochemical assessment of protein levels for the second type of glutaminase (GA) isoform, glutaminase gene encoding the liver-type isoforms (GLS2), did not detect substantial differences with regard to WT animals. The regional mRNA levels of GLS were determined by real time RT-PCR and did not show significant variations, except for prefrontal and motor cortex values which clearly diminished in KO mice. Total GA activity was also significantly reduced in prefrontal and motor cortex, but remained essentially unchanged in the hippocampus and rest of brain regions examined, suggesting activation of genetic compensatory mechanisms and/or post-translational modifications to compensate for KGA protein deficit. Remarkably, Golgi staining of hippocampal regions showed an altered morphology of glutamatergic pyramidal cells dendritic spines towards a less mature filopodia-like phenotype, as compared with WT littermates. This structural change correlated with a strong decrease of active matrix-metalloproteinase (MMP) 9 in cerebral cortex and hippocampus of KOLPA1 mice. Taken together, these results demonstrate that LPA signaling through LPA1 influence expression of the main isoenzyme of glutamate biosynthesis with strong repercussions on dendritic spines maturation, which may partially explain the cognitive and learning defects previously reported for this colony of KOLPA1 mice.
Collapse
Affiliation(s)
- Ana Peñalver
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - José A Campos-Sandoval
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - Eduardo Blanco
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de MálagaMálaga, Spain
| | - Carolina Cardona
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - Laura Castilla
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - Mercedes Martín-Rufián
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - Guillermo Estivill-Torrús
- Unidad de Gestión Clínica de Neurociencias, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de MálagaMálaga, Spain
| | - Raquel Sánchez-Varo
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Instituto de Investigación Biomédica de Málaga (IBIMA), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - Francisco J Alonso
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - Mercedes Pérez-Hernández
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de OctubreMadrid, Spain
| | - María I Colado
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de OctubreMadrid, Spain
| | - Antonia Gutiérrez
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Instituto de Investigación Biomédica de Málaga (IBIMA), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - Fernando Rodríguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de MálagaMálaga, Spain
| | - Javier Márquez
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de TeatinosMálaga, Spain
| |
Collapse
|
14
|
Hirayama-Kurogi M, Takizawa Y, Kunii Y, Matsumoto J, Wada A, Hino M, Akatsu H, Hashizume Y, Yamamoto S, Kondo T, Ito S, Tachikawa M, Niwa SI, Yabe H, Terasaki T, Setou M, Ohtsuki S. Downregulation of GNA13-ERK network in prefrontal cortex of schizophrenia brain identified by combined focused and targeted quantitative proteomics. J Proteomics 2017; 158:31-42. [DOI: 10.1016/j.jprot.2017.02.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 01/06/2023]
|
15
|
Glutaminolysis Was Induced by TGF-β1 through PP2Ac Regulated Raf-MEK-ERK Signaling in Endothelial Cells. PLoS One 2016; 11:e0162658. [PMID: 27612201 PMCID: PMC5017743 DOI: 10.1371/journal.pone.0162658] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 08/28/2016] [Indexed: 12/19/2022] Open
Abstract
Vascular endothelial cells can survive under hypoxic and inflammatory conditions by alterations of the cellular energy metabolism. In addition to high rates of glycolysis, glutaminolysis is another important way of providing the required energy to support cellular sprouting in such situations. However, the exact mechanism in which endothelial cells upregulate glutaminolysis remains unclear. Here we demonstrated that protein phosphatase 2A (PP2A)-mediated Raf-MEK-ERK signaling was involved in glutaminolysis in endothelial cells. Using models of human umbilical vein endothelial cells (HUVECs) treated with transforming growth factor-β1 (TGF-β1), we observed a dramatic induction in cellular glutamate levels accompanied by Raf-MEK-ERK activation. By addition of U0126, the specific inhibitor of MEK1/2, the expression of kidney-type glutaminase (KGA, a critical glutaminase in glutaminolysis) was significantly decreased. Moreover, inhibition of PP2A by okadaic acid (OA), a specific inhibitor of PP2A phosphatase activity or by depletion of its catalytic subunit (PP2Ac), led to a significant inactivation of Raf-MEK-ERK signaling and reduced glutaminolysis in endothelial cells. Taken together, these results indicated that PP2A-dependent Raf-MEK-ERK activation was involved in glutaminolysis and inhibition of PP2A signals was sufficient to block Raf-MEK-ERK pathway and reduced glutamine metabolism in endothelial cells.
Collapse
|
16
|
DAPK1 Signaling Pathways in Stroke: from Mechanisms to Therapies. Mol Neurobiol 2016; 54:4716-4722. [PMID: 27447806 PMCID: PMC5509806 DOI: 10.1007/s12035-016-0008-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 06/28/2016] [Indexed: 01/01/2023]
Abstract
Death-associated protein kinase 1 (DAPK1), a Ca2+/calmodulin (CaM)-dependent serine/threonine protein kinase, plays important roles in diverse apoptosis pathways not only in tumor suppression but also in neuronal cell death. The requirement of DAPK1 catalytic activity for its proposed cell functions and the elevation of catalytic activity of DAPK1 in injured neurons in models of neurological diseases, such as ischemia and epilepsy, validate that DAPK1 can be taken as a potential therapeutic target in these diseases. Recent studies show that DAPK1-NR2B, DAPK1-DANGER, DAPK1-p53, and DAPK1-Tau are currently known pathways in stroke-induced cell death, and blocking these cascades in an acute treatment effectively reduces neuronal loss. In this review, we focus on the role of DAPK1 in neuronal cell death after stroke. We hope to provide exhaustive summaries of relevant studies on DAPK1 signals involved in stroke damage. Therefore, disrupting DAPK1-relevant cell death pathway could be considered as a promising therapeutic approach in stroke.
Collapse
|
17
|
Shao WH, Chen JJ, Fan SH, Lei Y, Xu HB, Zhou J, Cheng PF, Yang YT, Rao CL, Wu B, Liu HP, Xie P. Combined Metabolomics and Proteomics Analysis of Major Depression in an Animal Model: Perturbed Energy Metabolism in the Chronic Mild Stressed Rat Cerebellum. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2016; 19:383-92. [PMID: 26134254 DOI: 10.1089/omi.2014.0164] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Major depressive disorder (MDD) is a highly prevalent, debilitating mental illness of importance for global health. However, its molecular pathophysiology remains poorly understood. Combined proteomics and metabolomics approaches should provide a comprehensive understanding of MDD's etiology. The present study reports novel "-omics" insights from a rodent model of MDD. Cerebellar samples from chronic mild stressed (CMS)-treated depressed rats and controls were compared with a focus on the differentially expressed proteins and metabolites using isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomics and gas chromotography/mass spectrometry (GC-MS) metabolomics techniques, respectively. The combined analyses found significant alterations associated with cerebellar energy metabolism, as indicated by (1) abnormal amino acid metabolism accompanied by corresponding metabolic enzymatic alterations and disturbed protein turnover, (2) increased glycolytic and tricarboxylic acid (TCA) cycle enzyme levels paralleled by changes in the concentrations of associated metabolites, and (3) perturbation of ATP biosynthesis through adenosine accompanied by perturbation of the mitochondrial respiratory chain. To the best of our knowledge, this study is the first to integrate proteomics and metabolomics analyses to examine the pathophysiological mechanism(s) underlying MDD in a CMS rodent model of depression. These results can offer important insights into the pathogenesis of MDD.
Collapse
Affiliation(s)
- Wei-hua Shao
- 1 Department of Respiratory Medicine, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Jian-jun Chen
- 3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Song-hua Fan
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Yang Lei
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Hong-bo Xu
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Jian Zhou
- 3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Peng-fei Cheng
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Yong-tao Yang
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Cheng-long Rao
- 3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Bo Wu
- 3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Hai-peng Liu
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Peng Xie
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| |
Collapse
|
18
|
Abstract
Mammalian glutaminases catalyze the stoichiometric conversion of L-glutamine to L-glutamate and ammonium ions. In brain, glutaminase is considered the prevailing pathway for synthesis of the neurotransmitter pool of glutamate. Besides neurotransmission, the products of glutaminase reaction also fulfill crucial roles in energy and metabolic homeostasis in mammalian brain. In the last years, new functional roles for brain glutaminases are being uncovered by using functional genomic and proteomic approaches. Glutaminases may act as multifunctional proteins able to perform different tasks: the discovery of multiple transcript variants in neurons and glial cells, novel extramitochondrial localizations, and isoform-specific proteininteracting partners strongly support possible moonlighting functions for these proteins. In this chapter, we present a critical account of essential works on brain glutaminase 80 years after its discovery. We will highlight the impact of recent findings and thoughts in the context of the glutamate/glutamine brain homeostasis.
Collapse
|
19
|
A pathway map of glutamate metabolism. J Cell Commun Signal 2015; 10:69-75. [PMID: 26635200 DOI: 10.1007/s12079-015-0315-5] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 11/25/2015] [Indexed: 10/22/2022] Open
Abstract
Glutamate metabolism plays a vital role in biosynthesis of nucleic acids and proteins. It is also associated with a number of different stress responses. Deficiency of enzymes involved in glutamate metabolism is associated with various disorders including gyrate atrophy, hyperammonemia, hemolytic anemia, γ-hydoxybutyric aciduria and 5-oxoprolinuria. Here, we present a pathway map of glutamate metabolism representing metabolic intermediates in the pathway, 107 regulator molecules, 9 interactors and 3 types of post-translational modifications. This pathway map provides detailed information about enzyme regulation, protein-enzyme interactions, post-translational modifications of enzymes and disorders due to enzyme deficiency. The information included in the map was based on published experimental evidence reported from mammalian systems.
Collapse
|
20
|
Hayakawa M, Itoh M, Ohta K, Li S, Ueda M, Wang MX, Nishida E, Islam S, Suzuki C, Ohzawa K, Kobori M, Inuzuka T, Nakagawa T. Quercetin reduces eIF2α phosphorylation by GADD34 induction. Neurobiol Aging 2015; 36:2509-18. [DOI: 10.1016/j.neurobiolaging.2015.05.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 04/28/2015] [Accepted: 05/10/2015] [Indexed: 01/11/2023]
|
21
|
Sun J, Pan C, Chew T, Liang F, Burmeister M, Low B. BNIP-H Recruits the Cholinergic Machinery to Neurite Terminals to Promote Acetylcholine Signaling and Neuritogenesis. Dev Cell 2015; 34:555-68. [DOI: 10.1016/j.devcel.2015.08.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 03/13/2015] [Accepted: 08/10/2015] [Indexed: 12/19/2022]
|
22
|
Abstract
Vocal production, which requires the generation and integration of laryngeal and respiratory motor patterns, can be impaired in dystonia, a disorder believed due to dysfunction of sensorimotor pathways in the central nervous system. Herein, we analyze vocal and respiratory abnormalities in the dystonic (dt) rat, a well-characterized model of generalized dystonia. The dt rat is a recessive mutant with haploinsufficiency of Atcay which encodes the neuronally restricted protein caytaxin. Olivocerebellar functional abnormalities are central to the dt rat's truncal and appendicular dystonia and could also contribute to vocal and respiratory abnormalities in this model system. Differences in vocal repertoire composition were found between homozygote and wild-type dt rat pups developing after 3 weeks of life. Those spectro-temporal differences were not paralleled by differences in vocal activity or maximum lung pressures during quiet breathing and vocalization. However, breathing rhythm was slower in homozygote pups. This slower breathing rhythm persisted into adulthood. Given that cerebellectomy eliminates truncal and appendicular dystonia in the dt rat, we hypothesize that the altered breathing patterns stem either from a disturbance in the maturation of respiratory pattern generators or from deficient extracerebellar caytaxin expression affecting normal respiratory pattern generation. The altered breathing rhythm associated with vocal changes in the murine model resembles aspects of vocal dysfunction that are seen in humans with sporadic dystonia.
Collapse
Affiliation(s)
- Tobias Riede
- Department of Physiology, Midwestern University, Glendale, Arizona
| | - Yu Zhao
- Department of Neurology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Mark S LeDoux
- Department of Neurology, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
23
|
LeDoux MS. Murine Models of Caytaxin Deficiency. Mov Disord 2015. [DOI: 10.1016/b978-0-12-405195-9.00025-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
24
|
Akamatsu R, Ishida-Kitagawa N, Aoyama T, Oka C, Kawaichi M. BNIP-2 binds phosphatidylserine, localizes to vesicles, and is transported by kinesin-1. Genes Cells 2014; 20:135-52. [PMID: 25472445 DOI: 10.1111/gtc.12209] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 10/19/2014] [Indexed: 11/30/2022]
Abstract
BNIP-2 shows high homology with the Cayman ataxia protein, caytaxin, which functions as a kinesin-1 adapter bridging cargos and kinesin light chains (KLCs). BNIP-2 is known to induce cell shape changes when over-expressed in culture cells, but its physiological functions are mostly unknown. BNIP-2 interacts with KLC through the conserved WED motif in the N-terminal region of BNIP-2. Interaction with KLC and transportation by kinesin-1 are essential for over-expressed BNIP-2 to elongate cells and induce cellular processes. Endogenous BNIP-2 localizes to the Golgi apparatus, early and recycling endosomes and mitochondria, aligned with microtubules, and moves at a speed compatible with kinesin-1 transportation. The CRAL-TRIO domain of BNIP-2 specifically interacts with phosphatidylserine, and the vesicular localization of BNIP-2 requires interaction with this phospholipid. BNIP-2 mutants which do not bind phosphatidylserine do not induce morphological changes in cells. These data show that similar to caytaxin, BNIP-2 is a kinesin-1 adapter involved in vesicular transportation in the cytoplasm and that association with cargos depends on interaction of the CRAL-TRIO domain with membrane phosphatidylserine.
Collapse
Affiliation(s)
- Rie Akamatsu
- Laboratory of Gene Function in Animals, Nara Institute of Science and Technology, 9816-5 Takayama, Ikoma, Nara, 630-0192, Japan
| | | | | | | | | |
Collapse
|
25
|
Yi P, Chew LL, Zhang Z, Ren H, Wang F, Cong X, Zheng L, Luo Y, Ouyang H, Low BC, Zhou YT. KIF5B transports BNIP-2 to regulate p38 mitogen-activated protein kinase activation and myoblast differentiation. Mol Biol Cell 2014; 26:29-42. [PMID: 25378581 PMCID: PMC4279227 DOI: 10.1091/mbc.e14-03-0797] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cdo bridges scaffold proteins BNIP-2 and JLP to activate p38MAPK during myoblast differentiation. KIF5B is a novel interacting partner of BNIP-2 and promotes myogenic differentiation. KIF5B-dependent transport of BNIP-2 is essential for its promyogenic effects. The Cdo-p38MAPK (p38 mitogen-activated protein kinase) signaling pathway plays important roles in regulating skeletal myogenesis. During myogenic differentiation, the cell surface receptor Cdo bridges scaffold proteins BNIP-2 and JLP and activates p38MAPK, but the spatial-temporal regulation of this process is largely unknown. We here report that KIF5B, the heavy chain of kinesin-1 motor, is a novel interacting partner of BNIP-2. Coimmunoprecipitation and far-Western study revealed that BNIP-2 directly interacted with the motor and tail domains of KIF5B via its BCH domain. By using a range of organelle markers and live microscopy, we determined the endosomal localization of BNIP-2 and revealed the microtubule-dependent anterograde transport of BNIP-2 in C2C12 cells. The anterograde transport of BNIP-2 was disrupted by a dominant-negative mutant of KIF5B. In addition, knockdown of KIF5B causes aberrant aggregation of BNIP-2, confirming that KIF5B is critical for the anterograde transport of BNIP-2 in cells. Gain- and loss-of-function experiments further showed that KIF5B modulates p38MAPK activity and in turn promotes myogenic differentiation. Of importance, the KIF5B-dependent anterograde transport of BNIP-2 is critical for its promyogenic effects. Our data reveal a novel role of KIF5B in the spatial regulation of Cdo–BNIP-2–p38MAPK signaling and disclose a previously unappreciated linkage between the intracellular transporting system and myogenesis regulation.
Collapse
Affiliation(s)
- Peng Yi
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Li Li Chew
- Department of Biological Sciences and Mechanobiology Institute, National University of Singapore, 117411 Singapore
| | - Ziwang Zhang
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hao Ren
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Feiya Wang
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoxia Cong
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Liling Zheng
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and
| | - Yan Luo
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and
| | - Hongwei Ouyang
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Boon Chuan Low
- Department of Biological Sciences and Mechanobiology Institute, National University of Singapore, 117411 Singapore
| | - Yi Ting Zhou
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
26
|
Glutamate metabolism and HIV-associated neurocognitive disorders. J Neurovirol 2014; 20:315-31. [PMID: 24867611 DOI: 10.1007/s13365-014-0258-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 03/14/2014] [Accepted: 05/09/2014] [Indexed: 12/21/2022]
Abstract
HIV-1 infection can lead to neurocognitive impairment collectively known as HIV-associated neurocognitive disorders (HAND). Although combined antiretroviral treatment (cART) has significantly ameliorated HIV's morbidity and mortality, persistent neuroinflammation and neurocognitive dysfunction continue. This review focuses on the current clinical and molecular evidence of the viral and host factors that influence glutamate-mediated neurotoxicity and neuropathogenesis as an important underlying mechanism during the course of HAND development. In addition, discusses potential pharmacological strategies targeting the glutamatergic system that may help prevent and improve neurological outcomes in HIV-1-infected subjects.
Collapse
|
27
|
Luna-Cancalon K, Sikora KM, Pappas SS, Singh V, Wulff H, Paulson HL, Burmeister M, Shakkottai VG. Alterations in cerebellar physiology are associated with a stiff-legged gait in Atcay(ji-hes) mice. Neurobiol Dis 2014; 67:140-8. [PMID: 24727095 DOI: 10.1016/j.nbd.2014.03.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 03/25/2014] [Accepted: 03/31/2014] [Indexed: 01/17/2023] Open
Abstract
Recent evidence suggests that dystonia, a movement disorder characterized by sustained involuntary muscle contractions, can be associated with cerebellar abnormalities. The basis for how functional changes in the cerebellum can cause dystonia is poorly understood. Here we identify alterations in physiology in Atcay(ji-hes) mice which in addition to ataxia, have an abnormal gait with hind limb extension and toe walking, reminiscent of human dystonic gait. No morphological abnormalities in the brain accompany the dystonia, but partial cerebellectomy causes resolution of the stiff-legged gait, suggesting that cerebellar dysfunction contributes to the dystonic gait of Atcay(ji-hes) mice. Recordings from Purkinje and deep cerebellar nuclear (DCN) neurons in acute brain slices were used to determine the physiological correlates of dystonia in the Atcay(ji-hes) mice. Approximately 50% of cerebellar Purkinje neurons fail to display the normal repetitive firing characteristic of these cells. In addition, DCN neurons exhibit increased intrinsic firing frequencies with a subset of neurons displaying bursts of action potentials. This increased intrinsic excitability of DCN neurons is accompanied by a reduction in after-hyperpolarization currents mediated by small-conductance calcium-activated potassium (SK) channels. An activator of SK channels reduces DCN neuron firing frequency in acute cerebellar slices and improves the dystonic gait of Atcay(ji-hes) mice. These results suggest that a combination of reduced Purkinje neuron activity and increased DCN intrinsic excitability can result in a combination of ataxia and a dystonia-like gait in mice.
Collapse
Affiliation(s)
| | - Kristine M Sikora
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Samuel S Pappas
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vikrant Singh
- Department of Pharmacology, University of California, Davis, CA 95616, USA
| | - Heike Wulff
- Department of Pharmacology, University of California, Davis, CA 95616, USA
| | - Henry L Paulson
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Margit Burmeister
- Molecular & Behavioral Neuroscience Institute, Departments of Psychiatry, Computational Medicine & Bioinformatics and Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
28
|
Glutaminase regulation in cancer cells: a druggable chain of events. Drug Discov Today 2013; 19:450-7. [PMID: 24140288 DOI: 10.1016/j.drudis.2013.10.008] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 09/09/2013] [Accepted: 10/08/2013] [Indexed: 12/21/2022]
Abstract
Metabolism is the process by which cells convert relatively simple extracellular nutrients into energy and building blocks necessary for their growth and survival. In cancer cells, metabolism is dramatically altered compared with normal cells. These alterations are known as the Warburg effect. One consequence of these changes is cellular addiction to glutamine. Because of this, in recent years the enzyme glutaminase has become a key target for small molecule therapeutic intervention. Like many oncotargets, however, glutaminase has a number of upstream partners that might offer additional druggable targets. This review summarizes the work from the current decade surrounding glutaminase and its regulation, and suggests strategies for therapeutic intervention in relevant cases.
Collapse
|
29
|
Márquez J, Cardona C, Campos-Sandoval JA, Peñalver A, Tosina M, Matés JM, Martín-Rufián M. Mammalian glutaminase isozymes in brain. Metab Brain Dis 2013; 28:133-7. [PMID: 23149879 DOI: 10.1007/s11011-012-9356-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 10/30/2012] [Indexed: 11/28/2022]
Abstract
Glutamine/glutamate homeostasis must be exquisitely regulated in mammalian brain and glutaminase (GA, E.C. 3.5.1.2) is one of the main enzymes involved. The products of GA reaction, glutamate and ammonia, are essential metabolites for energy and biosynthetic purposes but they are also hazardous compounds at concentrations beyond their normal physiological thresholds. The classical pattern of GA expression in mammals has been recently challenged by the discovery of novel transcript variants and protein isoforms. Furthermore, the interactome of brain GA is also starting to be uncovered adding a new level of regulatory complexity. GA may traffic in brain and unexpected locations, like cytosol and nucleus, have been found for GA isoforms. Finally, the expression of GA in glial cells has been reported and its potential implications in ammonia homeostasis are discussed.
Collapse
Affiliation(s)
- Javier Márquez
- Departamento de Biología Molecular y Bioquímica, Laboratorio de Química de Proteínas, Facultad de Ciencias, Universidad de Málaga, Málaga 29071, Spain.
| | | | | | | | | | | | | |
Collapse
|
30
|
Bmcc1s interacts with the phosphate-activated glutaminase in the brain. Biochimie 2012; 95:799-807. [PMID: 23246912 DOI: 10.1016/j.biochi.2012.11.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 11/26/2012] [Indexed: 11/24/2022]
Abstract
Bmcc1s, a brain-enriched short isoform of the BCH-domain containing molecule Bmcc1, has recently been shown to interact with the microtubule-associated protein MAP6 and to regulate cell morphology. Here we identified kidney-type glutaminase (KGA), the mitochondrial enzyme responsible for the conversion of glutamine to glutamate in neurons, as a novel partner of Bmcc1s. Co-immunoprecipitation experiments confirmed that Bmcc1s and KGA form a physiological complex in the brain, whereas binding and modeling studies showed that they interact with each other. Overexpression of Bmcc1s in mouse primary cortical neurons impaired proper mitochondrial targeting of KGA leading to its accumulation within the cytoplasm. Thus, Bmcc1s may control the trafficking of KGA to the mitochondria.
Collapse
|
31
|
Li S, Hayakawa-Yano Y, Itoh M, Ueda M, Ohta K, Suzuki Y, Mizuno A, Ohta E, Hida Y, Wang MX, Nakagawa T. Olfaxin as a novel Prune2 isoform predominantly expressed in olfactory system. Brain Res 2012; 1488:1-13. [DOI: 10.1016/j.brainres.2012.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 09/05/2012] [Accepted: 10/01/2012] [Indexed: 01/01/2023]
|
32
|
Sikora KM, Nosavanh LM, Kantheti P, Burmeister M, Hortsch M. Expression of Caytaxin protein in Cayman Ataxia mouse models correlates with phenotype severity. PLoS One 2012; 7:e50570. [PMID: 23226316 PMCID: PMC3511541 DOI: 10.1371/journal.pone.0050570] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 10/22/2012] [Indexed: 02/06/2023] Open
Abstract
Caytaxin is a highly-conserved protein, which is encoded by the Atcay/ATCAY gene. Mutations in Atcay/ATCAY have been identified as causative of cerebellar disorders such as the rare hereditary disease Cayman ataxia in humans, generalized dystonia in the dystonic (dt) rat, and marked motor defects in three ataxic mouse lines. While several lines of evidence suggest that Caytaxin plays a critical role in maintaining nervous system processes, the physiological function of Caytaxin has not been fully characterized. In the study presented here, we generated novel specific monoclonal antibodies against full-length Caytaxin to examine endogenous Caytaxin expression in wild type and Atcay mutant mouse lines. Caytaxin protein is absent from brain tissues in the two severely ataxic Atcayjit (jittery) and Atcayswd (sidewinder) mutant lines, and markedly decreased in the mildly ataxic/dystonic Atcayji-hes (hesitant) line, indicating a correlation between Caytaxin expression and disease severity. As the expression of wild type human Caytaxin in mutant sidewinder and jittery mice rescues the ataxic phenotype, Caytaxin’s physiological function appears to be conserved between the human and mouse orthologs. Across multiple species and in several neuronal cell lines Caytaxin is expressed as several protein isoforms, the two largest of which are caused by the usage of conserved methionine translation start sites. The work described in this manuscript presents an initial characterization of the Caytaxin protein and its expression in wild type and several mutant mouse models. Utilizing these animal models of human Cayman Ataxia will now allow an in-depth analysis to elucidate Caytaxin’s role in maintaining normal neuronal function.
Collapse
Affiliation(s)
- Kristine M. Sikora
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, United States of America
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - LaGina M. Nosavanh
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Prameela Kantheti
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Margit Burmeister
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, United States of America
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Psychiatry and Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Michael Hortsch
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
33
|
Pan CQ, Sudol M, Sheetz M, Low BC. Modularity and functional plasticity of scaffold proteins as p(l)acemakers in cell signaling. Cell Signal 2012; 24:2143-65. [PMID: 22743133 DOI: 10.1016/j.cellsig.2012.06.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 05/22/2012] [Accepted: 06/16/2012] [Indexed: 01/14/2023]
Abstract
Cells coordinate and integrate various functional modules that control their dynamics, intracellular trafficking, metabolism and gene expression. Such capacity is mediated by specific scaffold proteins that tether multiple components of signaling pathways at plasma membrane, Golgi apparatus, mitochondria, endoplasmic reticulum, nucleus and in more specialized subcellular structures such as focal adhesions, cell-cell junctions, endosomes, vesicles and synapses. Scaffold proteins act as "pacemakers" as well as "placemakers" that regulate the temporal, spatial and kinetic aspects of protein complex assembly by modulating the local concentrations, proximity, subcellular dispositions and biochemical properties of the target proteins through the intricate use of their modular protein domains. These regulatory mechanisms allow them to gate the specificity, integration and crosstalk of different signaling modules. In addition to acting as physical platforms for protein assembly, many professional scaffold proteins can also directly modify the properties of their targets while they themselves can be regulated by post-translational modifications and/or mechanical forces. Furthermore, multiple scaffold proteins can form alliances of higher-order regulatory networks. Here, we highlight the emerging themes of scaffold proteins by analyzing their common and distinctive mechanisms of action and regulation, which underlie their functional plasticity in cell signaling. Understanding these mechanisms in the context of space, time and force should have ramifications for human physiology and for developing new therapeutic approaches to control pathological states and diseases.
Collapse
Affiliation(s)
- Catherine Qiurong Pan
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Republic of Singapore.
| | | | | | | |
Collapse
|
34
|
Pan CQ, Low BC. Functional plasticity of the BNIP-2 and Cdc42GAP Homology (BCH) domain in cell signaling and cell dynamics. FEBS Lett 2012; 586:2674-91. [DOI: 10.1016/j.febslet.2012.04.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 04/16/2012] [Accepted: 04/16/2012] [Indexed: 10/28/2022]
|
35
|
Arama J, Boulay AC, Bosc C, Delphin C, Loew D, Rostaing P, Amigou E, Ezan P, Wingertsmann L, Guillaud L, Andrieux A, Giaume C, Cohen-Salmon M. Bmcc1s, a novel brain-isoform of Bmcc1, affects cell morphology by regulating MAP6/STOP functions. PLoS One 2012; 7:e35488. [PMID: 22523599 PMCID: PMC3327665 DOI: 10.1371/journal.pone.0035488] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 03/16/2012] [Indexed: 12/21/2022] Open
Abstract
The BCH (BNIP2 and Cdc42GAP Homology) domain-containing protein Bmcc1/Prune2 is highly enriched in the brain and is involved in the regulation of cytoskeleton dynamics and cell survival. However, the molecular mechanisms accounting for these functions are poorly defined. Here, we have identified Bmcc1s, a novel isoform of Bmcc1 predominantly expressed in the mouse brain. In primary cultures of astrocytes and neurons, Bmcc1s localized on intermediate filaments and microtubules and interacted directly with MAP6/STOP, a microtubule-binding protein responsible for microtubule cold stability. Bmcc1s overexpression inhibited MAP6-induced microtubule cold stability by displacing MAP6 away from microtubules. It also resulted in the formation of membrane protrusions for which MAP6 was a necessary cofactor of Bmcc1s. This study identifies Bmcc1s as a new MAP6 interacting protein able to modulate MAP6-induced microtubule cold stability. Moreover, it illustrates a novel mechanism by which Bmcc1 regulates cell morphology.
Collapse
Affiliation(s)
- Jessica Arama
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Anne-Cécile Boulay
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Christophe Bosc
- Equipe Physiopathologie du Cytosquelette, Institut National de la Santé et de la Recherche Médicale U836, Institut des Neurosciences, Université Joseph Fourier, Faculté de Médecine, Domaine de la Merci, La Tronche, France
| | - Christian Delphin
- Equipe Physiopathologie du Cytosquelette, Institut National de la Santé et de la Recherche Médicale U836, Institut des Neurosciences, Université Joseph Fourier, Faculté de Médecine, Domaine de la Merci, La Tronche, France
| | - Damarys Loew
- Institut Curie, Laboratory of Proteomic Mass Spectrometry, Paris, France
| | - Philippe Rostaing
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Institut National de la Santé et de la Recherche Médicale U1024, Paris, France
| | - Edwige Amigou
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Pascal Ezan
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Laure Wingertsmann
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Institut National de la Santé et de la Recherche Médicale U1024, Paris, France
| | - Laurent Guillaud
- Cell and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| | - Annie Andrieux
- Equipe Physiopathologie du Cytosquelette, Institut National de la Santé et de la Recherche Médicale U836, Institut des Neurosciences, Université Joseph Fourier, Faculté de Médecine, Domaine de la Merci, La Tronche, France
| | - Christian Giaume
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
| | - Martine Cohen-Salmon
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France
- University Pierre et Marie Curie, ED, N°158, Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France
- * E-mail:
| |
Collapse
|
36
|
Gupta AB, Wee LE, Zhou YT, Hortsch M, Low BC. Cross-species analyses identify the BNIP-2 and Cdc42GAP homology (BCH) domain as a distinct functional subclass of the CRAL_TRIO/Sec14 superfamily. PLoS One 2012; 7:e33863. [PMID: 22479462 PMCID: PMC3313917 DOI: 10.1371/journal.pone.0033863] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 02/18/2012] [Indexed: 11/19/2022] Open
Abstract
The CRAL_TRIO protein domain, which is unique to the Sec14 protein superfamily, binds to a diverse set of small lipophilic ligands. Similar domains are found in a range of different proteins including neurofibromatosis type-1, a Ras GTPase-activating Protein (RasGAP) and Rho guanine nucleotide exchange factors (RhoGEFs). Proteins containing this structural protein domain exhibit a low sequence similarity and ligand specificity while maintaining an overall characteristic three-dimensional structure. We have previously demonstrated that the BNIP-2 and Cdc42GAP Homology (BCH) protein domain, which shares a low sequence homology with the CRAL_TRIO domain, can serve as a regulatory scaffold that binds to Rho, RhoGEFs and RhoGAPs to control various cell signalling processes. In this work, we investigate 175 BCH domain-containing proteins from a wide range of different organisms. A phylogenetic analysis with ∼100 CRAL_TRIO and similar domains from eight representative species indicates a clear distinction of BCH-containing proteins as a novel subclass within the CRAL_TRIO/Sec14 superfamily. BCH-containing proteins contain a hallmark sequence motif R(R/K)h(R/K)(R/K)NL(R/K)xhhhhHPs (‘h’ is large and hydrophobic residue and ‘s’ is small and weekly polar residue) and can be further subdivided into three unique subtypes associated with BNIP-2-N, macro- and RhoGAP-type protein domains. A previously unknown group of genes encoding ‘BCH-only’ domains is also identified in plants and arthropod species. Based on an analysis of their gene-structure and their protein domain context we hypothesize that BCH domain-containing genes evolved through gene duplication, intron insertions and domain swapping events. Furthermore, we explore the point of divergence between BCH and CRAL-TRIO proteins in relation to their ability to bind small GTPases, GAPs and GEFs and lipid ligands. Our study suggests a need for a more extensive analysis of previously uncharacterized BCH, ‘BCH-like’ and CRAL_TRIO-containing proteins and their significance in regulating signaling events involving small GTPases.
Collapse
Affiliation(s)
- Anjali Bansal Gupta
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Liang En Wee
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Yi Ting Zhou
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Michael Hortsch
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Boon Chuan Low
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
37
|
Li S, Itoh M, Ohta K, Ueda M, Mizuno A, Ohta E, Hida Y, Wang MX, Takeuchi K, Nakagawa T. The expression and localization of Prune2 mRNA in the central nervous system. Neurosci Lett 2011; 503:208-14. [PMID: 21893162 DOI: 10.1016/j.neulet.2011.08.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 07/28/2011] [Accepted: 08/18/2011] [Indexed: 11/24/2022]
Abstract
A family of Bcl-2/adenovirus E1B 19kDa-interacting proteins (BNIPs) plays critical roles in several cellular processes such as cellular transformation, apoptosis, neuronal differentiation, and synaptic function, which are mediated by the BNIP2 and Cdc42GAP homology (BCH) domain. Prune homolog 2 (Drosophila) (PRUNE2) and its isoforms -C9orf65, BCH motif-containing molecule at the carboxyl terminal region 1 (BMCC1), and BNIP2 Extra Long (BNIPXL) - have been shown to be a susceptibility gene for Alzheimer's disease, a biomarker for leiomyosarcomas, a proapoptotic protein in neuronal cells, and an antagonist of cellular transformation, respectively. However, precise localization of PRUNE2 in the brain remains unclear. Here, we identified the distribution of Prune2 mRNA in the adult mouse brain. Prune2 mRNA is predominantly expressed in the neurons of the cranial nerve motor nuclei and the motor neurons of the spinal cord. The expression in the dorsal root ganglia (DRG) is consistent with the previously described reports. In addition, we observed the expression in another sensory neuron in the mesencephalic trigeminal nucleus. These results suggest that Prune2 may be functional in these restricted brain regions.
Collapse
Affiliation(s)
- Shimo Li
- Department of Neurobiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Piro RM, Molineris I, Ala U, Di Cunto F. Evaluation of candidate genes from orphan FEB and GEFS+ loci by analysis of human brain gene expression atlases. PLoS One 2011; 6:e23149. [PMID: 21858011 PMCID: PMC3157479 DOI: 10.1371/journal.pone.0023149] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 07/07/2011] [Indexed: 12/19/2022] Open
Abstract
Febrile seizures, or febrile convulsions (FEB), represent the most common form of childhood seizures and are believed to be influenced by variations in several susceptibility genes. Most of the associated loci, however, remain ‘orphan’, i.e. the susceptibility genes they contain still remain to be identified. Further orphan loci have been mapped for a related disorder, genetic (generalized) epilepsy with febrile seizures plus (GEFS+). We show that both spatially mapped and ‘traditional’ gene expression data from the human brain can be successfully employed to predict the most promising candidate genes for FEB and GEFS+, apply our prediction method to the remaining orphan loci and discuss the validity of the predictions. For several of the orphan FEB/GEFS+ loci we propose excellent, and not always obvious, candidates for mutation screening in order to aid in gaining a better understanding of the genetic origin of the susceptibility to seizures.
Collapse
Affiliation(s)
- Rosario M Piro
- Molecular Biotechnology Center and Department of Genetics, Biology and Biochemistry, University of Torino, Torino, Italy.
| | | | | | | |
Collapse
|
39
|
LeDoux MS. Animal models of dystonia: Lessons from a mutant rat. Neurobiol Dis 2011; 42:152-61. [PMID: 21081162 PMCID: PMC3171987 DOI: 10.1016/j.nbd.2010.11.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 10/15/2010] [Accepted: 11/09/2010] [Indexed: 01/25/2023] Open
Abstract
Dystonia is a motor sign characterized by involuntary muscle contractions which produce abnormal postures. Genetic factors contribute significantly to primary dystonia. In comparison, secondary dystonia can be caused by a wide variety of metabolic, structural, infectious, toxic and inflammatory insults to the nervous system. Although classically ascribed to dysfunction of the basal ganglia, studies of diverse animal models have pointed out that dystonia is a network disorder with important contributions from abnormal olivocerebellar signaling. In particular, work with the dystonic (dt) rat has engendered dramatic paradigm shifts in dystonia research. The dt rat manifests generalized dystonia caused by deficiency of the neuronally restricted protein caytaxin. Electrophysiological and biochemical studies have shown that defects at the climbing fiber-Purkinje cell synapse in the dt rat lead to abnormal bursting firing patterns in the cerebellar nuclei, which increases linearly with postnatal age. In a general sense, the dt rat has shown the scientific and clinical communities that dystonia can arise from dysfunctional cerebellar cortex. Furthermore, work with the dt rat has provided evidence that dystonia (1) is a neurodevelopmental network disorder and (2) can be driven by abnormal cerebellar output. In large part, work with other animal models has expanded upon studies in the dt rat and shown that primary dystonia is a multi-nodal network disorder associated with defective sensorimotor integration. In addition, experiments in genetically engineered models have been used to examine the underlying cellular pathologies that drive primary dystonia. This article is part of a Special Issue entitled "Advances in dystonia".
Collapse
Affiliation(s)
- Mark S LeDoux
- University of Tennessee Health Science Center, Department of Neurology, 855 Monroe Avenue, Link Building, Suite 415, Memphis, TN 38163, USA.
| |
Collapse
|
40
|
Cayman Ataxia-Related Protein is a Presynapse-Specific Caspase-3 Substrate. Neurochem Res 2011; 36:1304-13. [DOI: 10.1007/s11064-011-0430-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2011] [Indexed: 11/25/2022]
|
41
|
Zhou YT, Chew LL, Lin SC, Low BC. The BNIP-2 and Cdc42GAP homology (BCH) domain of p50RhoGAP/Cdc42GAP sequesters RhoA from inactivation by the adjacent GTPase-activating protein domain. Mol Biol Cell 2010; 21:3232-46. [PMID: 20660160 PMCID: PMC2938388 DOI: 10.1091/mbc.e09-05-0408] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The BNIP-2 and Cdc42GAP Homology (BCH) domain from p50RhoGAP sequesters RhoA from inactivation by the adjacent GAP domain and it confers unique Rho-binding profile from that of GAP domain. This suppression is further augmented by an intramolecular interaction, adding to a new paradigm for regulating p50RhoGAP signaling. The BNIP-2 and Cdc42GAP homology (BCH) domain is a novel regulator for Rho GTPases, but its impact on p50-Rho GTPase-activating protein (p50RhoGAP or Cdc42GAP) in cells remains elusive. Here we show that deletion of the BCH domain from p50RhoGAP enhanced its GAP activity and caused drastic cell rounding. Introducing constitutively active RhoA or inactivating GAP domain blocked such effect, whereas replacing the BCH domain with endosome-targeting SNX3 excluded requirement of endosomal localization in regulating the GAP activity. Substitution with homologous BCH domain from Schizosaccharomyces pombe, which does not bind mammalian RhoA, also led to complete loss of suppression. Interestingly, the p50RhoGAP BCH domain only targeted RhoA, but not Cdc42 or Rac1, and it was unable to distinguish between GDP and the GTP-bound form of RhoA. Further mutagenesis revealed a RhoA-binding motif (residues 85-120), which when deleted, significantly reduced BCH inhibition on GAP-mediated cell rounding, whereas its full suppression also required an intramolecular interaction motif (residues 169-197). Therefore, BCH domain serves as a local modulator in cis to sequester RhoA from inactivation by the adjacent GAP domain, adding to a new paradigm for regulating p50RhoGAP signaling.
Collapse
Affiliation(s)
- Yi Ting Zhou
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore 117543, Republic of Singapore.
| | | | | | | |
Collapse
|
42
|
Abstract
AbstractGlutaminase is considered as the main glutamate producer enzyme in brain. Consequently, the enzyme is essential for both glutamatergic and gabaergic transmissions. Glutamine-derived glutamate and ammonia, the products of glutaminase reaction, fulfill crucial roles in energy metabolism and in the biosynthesis of basic metabolites, such as GABA, proteins and glutathione. However, glutamate and ammonia are also hazardous compounds and danger lurks in their generation beyond normal physiological thresholds; hence, glutaminase activity must be carefully regulated in the mammalian brain. The differential distribution and regulation of glutaminase are key factors to modulate the metabolism of glutamate and glutamine in brain. The discovery of novel isoenzymes, protein interacting partners and subcellular localizations indicate new functions for brain glutaminase. In this short review, we summarize recent findings that point consistently towards glutaminase as a multifaceted protein able to perform different tasks. Finally, we will highlight the involvement of glutaminase in pathological states and its consideration as a potential therapeutic target.
Collapse
|
43
|
Filiou MD, Bisle B, Reckow S, Teplytska L, Maccarrone G, Turck CW. Profiling of mouse synaptosome proteome and phosphoproteome by IEF. Electrophoresis 2010; 31:1294-301. [DOI: 10.1002/elps.200900647] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
44
|
Aoyama T, Hata S, Nakao T, Tanigawa Y, Oka C, Kawaichi M. Cayman ataxia protein caytaxin is transported by kinesin along neurites through binding to kinesin light chains. J Cell Sci 2009; 122:4177-85. [DOI: 10.1242/jcs.048579] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Deficiency of caytaxin results in hereditary ataxia or dystonia in humans, mice and rats. Our yeast two-hybrid screen identified kinesin light chains (KLCs) as caytaxin-binding proteins. The tetratricopeptide-repeat region of KLC1 recognizes the ELEWED sequence (amino acids 115-120) of caytaxin. This motif is conserved among BNIP-2 family members and other KLC-interacting kinesin cargo proteins such as calsyntenins. Caytaxin associates with kinesin heavy chains (KHCs) indirectly by binding to KLCs, suggesting that caytaxin binds to the tetrameric kinesin molecule. In cultured hippocampal neurons, we found that caytaxin is distributed in both axons and dendrites in punctate patterns, and it colocalizes with microtubules and KHC. GFP-caytaxin expressed in hippocampal neurons is transported at a speed (∼1 μm/second) compatible with kinesin movement. Inhibition of kinesin-1 by dominant-negative KHC decreases the accumulation of caytaxin in the growth cone. Caytaxin puncta do not coincide with vesicles containing known kinesin cargos such as APP or JIP-1. A part of caytaxin, however, colocalizes with mitochondria and suppression of caytaxin expression by RNAi redistributes mitochondria away from the distal ends of neurites. These data indicate that caytaxin binds to kinesin-1 and functions as an adaptor that mediates intracellular transport of specific cargos, one of which is the mitochondrion.
Collapse
Affiliation(s)
- Takane Aoyama
- Division of Gene Function in Animals, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Suguru Hata
- Division of Gene Function in Animals, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Takeshi Nakao
- Division of Gene Function in Animals, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Yuka Tanigawa
- Division of Gene Function in Animals, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Chio Oka
- Division of Gene Function in Animals, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Masashi Kawaichi
- Division of Gene Function in Animals, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| |
Collapse
|
45
|
|
46
|
Concepcion D, Flores-García L, Hamilton BA. Multipotent genetic suppression of retrotransposon-induced mutations by Nxf1 through fine-tuning of alternative splicing. PLoS Genet 2009; 5:e1000484. [PMID: 19436707 PMCID: PMC2674570 DOI: 10.1371/journal.pgen.1000484] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Accepted: 04/17/2009] [Indexed: 11/18/2022] Open
Abstract
Cellular gene expression machinery has coevolved with molecular parasites, such as viruses and transposons, which rely on host cells for their expression and reproduction. We previously reported that a wild-derived allele of mouse Nxf1 (Tap), a key component of the host mRNA nuclear export machinery, suppresses two endogenous retrovirus-induced mutations and shows suggestive evidence of positive selection. Here we show that Nxf1CAST suppresses a specific and frequent class of intracisternal A particle (IAP)-induced mutations, including Ap3d1mh2J, a model for Hermansky-Pudlak syndrome, and Atcayhes, an orthologous gene model for Cayman ataxia, among others. The molecular phenotype of suppression includes ∼two-fold increase in the level of correctly-spliced mRNA and a decrease in mutant-specific, alternatively-processed RNA accumulating from the inserted allele. Insertional mutations involving ETn and LINE elements are not suppressed, demonstrating a high degree of specificity to this suppression mechanism. These results implicate Nxf1 in some instances of pre-mRNA processing, demonstrate the useful range of Nxf1CAST alleles for manipulating existing mouse models of disease, and specifically imply a low functional threshold for therapeutic benefit in Cayman ataxia. Retroviruses and transposable elements are molecular parasites that integrate into the host genome and require host cell machinery for gene expression, replication and dissemination. Integrating elements can alter the expression of nearby host genes through both transcriptional and post-transcriptional mechanisms. Components of the host cell machinery that can adapt to favor genetic programs of the host cell over those of the parasite may afford one level of innate immunity. In laboratory mice, endogenous retroviruses are virus-derived mobile elements that account for many spontaneous mutations. A frequent class involves retrotransposition into introns of genes in the transcriptional sense orientation, which alters host gene pre-mRNA splicing. Here we show that for the intracisternal A particle (IAP) family of endogenous retroviruses, an allele of the canonical mRNA export factor Nxf1 found in wild Asiatic mice (Mus musculus castaneus) suppresses most insertions of this class (six of seven tested). To our knowledge, these results make Nxf1 the most broadly interacting modifier gene yet documented in this well-studied species. These results have significant implications for manipulating gene expression in mouse models of disease, the role of Nxf1 in pre-mRNA processing and in the dynamic range for therapeutic intervention in Cayman ataxia.
Collapse
Affiliation(s)
- Dorothy Concepcion
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, California, United States of America
- Department of Cellular and Molecular Medicine, University of California San Diego School of Medicine, La Jolla, California, United States of America
- Rebecca and John Moores UCSD Cancer Center, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Lisbeth Flores-García
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, California, United States of America
- Department of Cellular and Molecular Medicine, University of California San Diego School of Medicine, La Jolla, California, United States of America
- Rebecca and John Moores UCSD Cancer Center, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Bruce A. Hamilton
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, California, United States of America
- Department of Cellular and Molecular Medicine, University of California San Diego School of Medicine, La Jolla, California, United States of America
- Rebecca and John Moores UCSD Cancer Center, University of California San Diego School of Medicine, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
47
|
New insights into brain glutaminases: beyond their role on glutamatergic transmission. Neurochem Int 2009; 55:64-70. [PMID: 19428808 DOI: 10.1016/j.neuint.2009.02.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2008] [Revised: 02/18/2009] [Accepted: 02/27/2009] [Indexed: 01/29/2023]
Abstract
The synthesis of glutamate in brain must be exquisitely regulated because of its harmful potential giving rise to excitotoxic damage. In this sense, a stringent control based on multiple regulatory mechanisms should be expected to be exhibited by the biosynthetic enzymes responsible of glutamate generation, to assure that glutamate is only synthesized at the right place and at the right time. Glutaminase is considered as the main glutamate-producer enzyme in brain. Recently, novel glutaminase isoforms and extramitochondrial locations for these proteins have been discovered in the brain of mammals: identifying the function of each isozyme is essential for understanding the role of glutaminases in cerebral function. In addition, the interactome of glutaminases is starting to be uncovered adding a new level of regulatory complexity with important functional consequences, including selective and regulated targeting to concrete cellular locations. Finally, recent progress has identified glutaminase to be also present in astrocytes which precludes its classical consideration as a neuron-specific enzyme.
Collapse
|
48
|
Buschdorf JP, Chew LL, Soh UJK, Liou YC, Low BC. Nerve growth factor stimulates interaction of Cayman ataxia protein BNIP-H/Caytaxin with peptidyl-prolyl isomerase Pin1 in differentiating neurons. PLoS One 2008; 3:e2686. [PMID: 18628984 PMCID: PMC2442193 DOI: 10.1371/journal.pone.0002686] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Accepted: 06/08/2008] [Indexed: 11/18/2022] Open
Abstract
Mutations in ATCAY that encodes the brain-specific protein BNIP-H (or Caytaxin) lead to Cayman cerebellar ataxia. BNIP-H binds to glutaminase, a neurotransmitter-producing enzyme, and affects its activity and intracellular localization. Here we describe the identification and characterization of the binding between BNIP-H and Pin1, a peptidyl-prolyl cis/trans isomerase. BNIP-H interacted with Pin1 after nerve growth factor-stimulation and they co-localized in the neurites and cytosol of differentiating pheochromocytoma PC12 cells and the embryonic carcinoma P19 cells. Deletional mutagenesis revealed two cryptic binding sites within the C-terminus of BNIP-H such that single point mutants affecting the WW domain of Pin1 completely abolished their binding. Although these two sites do not contain any of the canonical Pin1-binding motifs they showed differential binding profiles to Pin1 WW domain mutants S16E, S16A and W34A, and the catalytically inert C113A of its isomerase domain. Furthermore, their direct interaction would occur only upon disrupting the ability of BNIP-H to form an intramolecular interaction by two similar regions. Furthermore, expression of Pin1 disrupted the BNIP-H/glutaminase complex formation in PC12 cells under nerve growth factor-stimulation. These results indicate that nerve growth factor may stimulate the interaction of BNIP-H with Pin1 by releasing its intramolecular inhibition. Such a mechanism could provide a post-translational regulation on the cellular activity of BNIP-H during neuronal differentiation.
Collapse
Affiliation(s)
- Jan Paul Buschdorf
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Republic of Singapore
| | | | | | | | | |
Collapse
|
49
|
Soh UJK, Low BC. BNIP2 extra long inhibits RhoA and cellular transformation by Lbc RhoGEF via its BCH domain. J Cell Sci 2008; 121:1739-49. [DOI: 10.1242/jcs.021774] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Increased expression of BCH-motif-containing molecule at the C-terminal region 1 (BMCC1) correlates with a favourable prognosis in neuroblastoma, but the underlying mechanism remains unknown. We here isolated BNIPXL (BNIP2 Extra Long) as a single contig of the extended, in-vitro-assembled BMCC1. Here, we show that in addition to homophilic interactions, the BNIP2 and Cdc42GAP homology (BCH) domain of BNIPXL interacts with specific conformers of RhoA and also mediates association with the catalytic DH-PH domains of Lbc, a RhoA-specific guanine nucleotide exchange factor (RhoGEF). BNIPXL does not recognize the constitutive active G14V and Q63L mutants of RhoA but targets the fast-cycling F30L and the dominant-negative T19N mutants. A second region at the N-terminus of BNIPXL also targets the proline-rich region of Lbc. Whereas overexpression of BNIPXL reduces active RhoA levels, knockdown of BNIPXL expression has the reverse effect. Consequently, BNIPXL inhibits Lbc-induced oncogenic transformation. Interestingly, BNIPXL can also interact with RhoC, but not with RhoB. Given the importance of RhoA and RhoGEF signaling in tumorigenesis, BNIPXL could suppress cellular transformation by preventing sustained Rho activation in concert with restricting RhoA and Lbc binding via its BCH domain. This could provide a general mechanism for regulating RhoGEFs and their target GTPases.
Collapse
Affiliation(s)
- Unice J. K. Soh
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Republic of Singapore
| | - Boon Chuan Low
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Republic of Singapore
| |
Collapse
|
50
|
Breakefield XO, Blood AJ, Li Y, Hallett M, Hanson PI, Standaert DG. The pathophysiological basis of dystonias. Nat Rev Neurosci 2008; 9:222-34. [PMID: 18285800 DOI: 10.1038/nrn2337] [Citation(s) in RCA: 327] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dystonias comprise a group of movement disorders that are characterized by involuntary movements and postures. Insight into the nature of neuronal dysfunction has been provided by the identification of genes responsible for primary dystonias, the characterization of animal models and functional evaluations and in vivo brain imaging of patients with dystonia. The data suggest that alterations in neuronal development and communication within the brain create a susceptible substratum for dystonia. Although there is no overt neurodegeneration in most forms of dystonia, there are functional and microstructural brain alterations. Dystonia offers a window into the mechanisms whereby subtle changes in neuronal function, particularly in sensorimotor circuits that are associated with motor learning and memory, can corrupt normal coordination and lead to a disabling motor disorder.
Collapse
Affiliation(s)
- Xandra O Breakefield
- Department of Neurology and Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA.
| | | | | | | | | | | |
Collapse
|