1
|
Lange KR, Rasheed N, Su X, Diaz-Rubio ME, Firestein BL. Valacyclovir and Acyclovir Are Substrates of the Guanine Deaminase Cytosolic PSD-95 Interactor (Cypin). Proteins 2024. [PMID: 39210666 DOI: 10.1002/prot.26740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/27/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
Valacyclovir, enzymatically hydrolyzed in the body to acyclovir, is a guanine-based nucleoside analog commonly prescribed as an antiviral therapy. Previous reports suggest that guanosine analogs bind to guanine deaminase; however, it is unclear whether they act as inhibitors or substrates. Data from our laboratory suggest that inhibition of guanine deaminase by small molecules attenuates spinal cord injury-induced neuropathic pain. Here, we examine whether the guanosine analogs valacyclovir and acyclovir are deaminated by cypin (cytosolic PSD-95 interactor), the major guanine deaminase in the body, or if they act as cypin inhibitors. Using purified Rattus norvegicus cypin, we use NADH-coupled assay to confirm deamination of valacyclovir and determined Michaelis-Menten constants. Subsequently, we use tryptophan fluorescence quenching assay to calculate dissociation constants for valacyclovir and acyclovir and find that inclusion of the valine motif in valacyclovir increases affinity for cypin compared to acyclovir. To our knowledge, neither Km nor KD values for cypin has been previously reported for either compound. We use Amplex Red assay and demonstrate that both valacyclovir and acyclovir are cypin substrates and that their metabolites are further processed by xanthine oxidase and uricase. Using molecular dynamics simulations, we demonstrate that an alpha helix near the active site is displaced when valacyclovir binds to cypin. Furthermore, we used LC-MS-based assay to directly confirm deamination of valacyclovir by cypin. Taken together, our results demonstrate a novel role for cypin in deamination of valacyclovir and acyclovir and suggest that therapeutics based on purine structures may be inactivated by cypin, decreasing inhibitory efficacy.
Collapse
Affiliation(s)
- Keith R Lange
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
- Graduate Program in Biochemistry, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Noor Rasheed
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Xiaoyang Su
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
- Rutgers Metabolomics Shared Resource, Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - M Elena Diaz-Rubio
- Rutgers Metabolomics Shared Resource, Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| |
Collapse
|
2
|
Akere MT, Zajac KK, Bretz JD, Madhavaram AR, Horton AC, Schiefer IT. Real-Time Analysis of Neuronal Cell Cultures for CNS Drug Discovery. Brain Sci 2024; 14:770. [PMID: 39199464 PMCID: PMC11352746 DOI: 10.3390/brainsci14080770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/23/2024] [Accepted: 07/27/2024] [Indexed: 09/01/2024] Open
Abstract
The ability to screen for agents that can promote the development and/or maintenance of neuronal networks creates opportunities for the discovery of novel agents for the treatment of central nervous system (CNS) disorders. Over the past 10 years, advances in robotics, artificial intelligence, and machine learning have paved the way for the improved implementation of live-cell imaging systems for drug discovery. These instruments have revolutionized our ability to quickly and accurately acquire large standardized datasets when studying complex cellular phenomena in real-time. This is particularly useful in the field of neuroscience because real-time analysis can allow efficient monitoring of the development, maturation, and conservation of neuronal networks by measuring neurite length. Unfortunately, due to the relative infancy of this type of analysis, standard practices for data acquisition and processing are lacking, and there is no standardized format for reporting the vast quantities of data generated by live-cell imaging systems. This paper reviews the current state of live-cell imaging instruments, with a focus on the most commonly used equipment (IncuCyte systems). We provide an in-depth analysis of the experimental conditions reported in publications utilizing these systems, particularly with regard to studying neurite outgrowth. This analysis sheds light on trends and patterns that will enhance the use of live-cell imaging instruments in CNS drug discovery.
Collapse
Affiliation(s)
- Millicent T. Akere
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (M.T.A.); (K.K.Z.); (J.D.B.); (A.R.M.); (A.C.H.)
| | - Kelsee K. Zajac
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (M.T.A.); (K.K.Z.); (J.D.B.); (A.R.M.); (A.C.H.)
| | - James D. Bretz
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (M.T.A.); (K.K.Z.); (J.D.B.); (A.R.M.); (A.C.H.)
| | - Anvitha R. Madhavaram
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (M.T.A.); (K.K.Z.); (J.D.B.); (A.R.M.); (A.C.H.)
| | - Austin C. Horton
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (M.T.A.); (K.K.Z.); (J.D.B.); (A.R.M.); (A.C.H.)
| | - Isaac T. Schiefer
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (M.T.A.); (K.K.Z.); (J.D.B.); (A.R.M.); (A.C.H.)
- Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
3
|
Singh NK, Gandu SR, Li L, Ni L, Acioglu C, Mirabelli E, Hiester LL, Elkabes S, Firestein BL. Cypin Inhibition as a Therapeutic Approach to Treat Spinal Cord Injury-Induced Mechanical Pain. eNeuro 2024; 11:ENEURO.0451-23.2024. [PMID: 38302457 PMCID: PMC10875717 DOI: 10.1523/eneuro.0451-23.2024] [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: 10/30/2023] [Revised: 12/24/2023] [Accepted: 01/04/2024] [Indexed: 02/03/2024] Open
Abstract
Cypin (cytosolic postsynaptic density protein 95 interactor) is the primary guanine deaminase in the central nervous system (CNS), promoting the metabolism of guanine to xanthine, an important reaction in the purine salvage pathway. Activation of the purine salvage pathway leads to the production of uric acid (UA). UA has paradoxical effects, specifically in the context of CNS injury as it confers neuroprotection, but it also promotes pain. Since neuropathic pain is a comorbidity associated with spinal cord injury (SCI), we postulated that small molecule cypin inhibitor B9 treatment could attenuate SCI-induced neuropathic pain, potentially by interfering with UA production. However, we also considered that this treatment could hinder the neuroprotective effects of UA and, in doing so, exacerbate SCI outcomes. To address our hypothesis, we induced a moderate midthoracic contusion SCI in female mice and assessed whether transient intrathecal administration of B9, starting at 1 d postinjury (dpi) until 7 dpi, attenuates mechanical pain in hindlimbs at 3 weeks pi. We also evaluated the effects of B9 on the spontaneous recovery of locomotor function. We found that B9 alleviates mechanical pain but does not affect locomotor function. Importantly, B9 does not exacerbate lesion volume at the epicenter. In accordance with these findings, B9 does not aggravate glutamate-induced excitotoxic death of SC neurons in vitro. Moreover, SCI-induced increased astrocyte reactivity at the glial scar is not altered by B9 treatment. Our data suggest that B9 treatment reduces mechanical pain without exerting major detrimental effects following SCI.
Collapse
Affiliation(s)
- Nisha K Singh
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854
- Molecular Biosciences Graduate Program, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854
| | - Srinivasa R Gandu
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854
- Molecular Biosciences Graduate Program, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854
| | - Lun Li
- Department of Neurosurgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey 07101
| | - Li Ni
- Department of Neurosurgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey 07101
| | - Cigdem Acioglu
- Department of Neurosurgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey 07101
| | - Ersilia Mirabelli
- Department of Neurosurgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey 07101
| | - Liam L Hiester
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854
| | - Stella Elkabes
- Department of Neurosurgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey 07101
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854
| |
Collapse
|
4
|
Della Rosa G, Di Buduo CA, Balduini A. Unraveling the hormonal pathway to megakaryocyte well-being. J Thromb Haemost 2023; 21:3078-3081. [PMID: 37858524 DOI: 10.1016/j.jtha.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 10/21/2023]
Affiliation(s)
| | | | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA.
| |
Collapse
|
5
|
Grodzielski M, Cidlowski JA. Glucocorticoids regulate thrombopoiesis by remodeling the megakaryocyte transcriptome. J Thromb Haemost 2023; 21:3207-3223. [PMID: 37336437 PMCID: PMC10592358 DOI: 10.1016/j.jtha.2023.06.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/18/2023] [Accepted: 06/07/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND Glucocorticoids are widely known for their immunomodulatory action. Their synthetic analogs are used to treat several autoimmune diseases, including immune thrombocytopenia. However, their efficacy and mechanisms of action in immune thrombocytopenia are not fully understood. OBJECTIVES To investigate the mechanism of glucocorticoid actions on platelet production. METHODS The actions of glucocorticoids on platelet production were studied combining in vivo, ex vivo and in vitro approaches. RESULTS Dexamethasone reduced bleeding in mice and rapidly increased circulating young platelet counts. In vitro glucocorticoid treatment stimulated proplatelet formation by megakaryocytes and platelet-like particle release. This effect was blocked by glucocorticoid receptor antagonist RU486, indicating a glucocorticoid receptor-dependent mechanism. Genome-wide analysis revealed that dexamethasone regulates the expression of >1000 genes related to numerous cellular functions, including predominant cytoplasm and cytoskeleton reorganization. Dexamethasone and other glucocorticoids induced the expression of Gda (the gene encoding guanine deaminase), which has been reported to have a role in dendrite development. Inhibition of guanine deaminase enzymatic activity blocked dexamethasone stimulation of proplatelet formation, implicating a critical role for this enzyme in glucocorticoid-mediated platelet production. CONCLUSION Our findings identify glucocorticoids as new regulators of thrombopoiesis.
Collapse
Affiliation(s)
- Matías Grodzielski
- Molecular Endocrinology Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - John A Cidlowski
- Molecular Endocrinology Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA.
| |
Collapse
|
6
|
Siddiq MM, Toro CA, Johnson NP, Hansen J, Xiong Y, Mellado W, Tolentino RE, Johnson K, Jayaraman G, Suhail Z, Harlow L, Dai J, Beaumont KG, Sebra R, Willis DE, Cardozo CP, Iyengar R. Spinal cord injury regulates circular RNA expression in axons. Front Mol Neurosci 2023; 16:1183315. [PMID: 37692100 PMCID: PMC10483835 DOI: 10.3389/fnmol.2023.1183315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/04/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction Neurons transport mRNA and translational machinery to axons for local translation. After spinal cord injury (SCI), de novo translation is assumed to enable neurorepair. Knowledge of the identity of axonal mRNAs that participate in neurorepair after SCI is limited. We sought to identify and understand how axonal RNAs play a role in axonal regeneration. Methods We obtained preparations enriched in axonal mRNAs from control and SCI rats by digesting spinal cord tissue with cold-active protease (CAP). The digested samples were then centrifuged to obtain a supernatant that was used to identify mRNA expression. We identified differentially expressed genes (DEGS) after SCI and mapped them to various biological processes. We validated the DEGs by RT-qPCR and RNA-scope. Results The supernatant fraction was highly enriched for mRNA from axons. Using Gene Ontology, the second most significant pathway for all DEGs was axonogenesis. Among the DEGs was Rims2, which is predominately a circular RNA (circRNA) in the CNS. We show that Rims2 RNA within spinal cord axons is circular. We found an additional 200 putative circRNAs in the axonal-enriched fraction. Knockdown in primary rat cortical neurons of the RNA editing enzyme ADAR1, which inhibits formation of circRNAs, significantly increased axonal outgrowth and increased the expression of circRims2. Using Rims2 as a prototype we used Circular RNA Interactome to predict miRNAs that bind to circRims2 also bind to the 3'UTR of GAP-43, PTEN or CREB1, all known regulators of axonal outgrowth. Axonally-translated GAP-43 supports axonal elongation and we detect GAP-43 mRNA in the rat axons by RNAscope. Discussion By enriching for axonal RNA, we detect SCI induced DEGs, including circRNA such as Rims2. Ablation of ADAR1, the enzyme that regulates circRNA formation, promotes axonal outgrowth of cortical neurons. We developed a pathway model using Circular RNA Interactome that indicates that Rims2 through miRNAs can regulate the axonal translation GAP-43 to regulate axonal regeneration. We conclude that axonal regulatory pathways will play a role in neurorepair.
Collapse
Affiliation(s)
- Mustafa M. Siddiq
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Carlos A. Toro
- Spinal Cord Damage Research Center, James J. Peters VA Medical Center, Bronx, NY, United States
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nicholas P. Johnson
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jens Hansen
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Yuguang Xiong
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Rosa E. Tolentino
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kaitlin Johnson
- Spinal Cord Damage Research Center, James J. Peters VA Medical Center, Bronx, NY, United States
| | - Gomathi Jayaraman
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Zaara Suhail
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Lauren Harlow
- Spinal Cord Damage Research Center, James J. Peters VA Medical Center, Bronx, NY, United States
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jinye Dai
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kristin G. Beaumont
- Department of Genetics and Genomic Studies, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Icahn Genomics Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Robert Sebra
- Department of Genetics and Genomic Studies, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Icahn Genomics Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Dianna E. Willis
- Burke Neurological Institute, White Plains, NY, United States
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Christopher P. Cardozo
- Spinal Cord Damage Research Center, James J. Peters VA Medical Center, Bronx, NY, United States
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ravi Iyengar
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
7
|
Liu J, Kong W, Liu Y, Ma Q, Shao Q, Zeng L, Chao Y, Song X, Zhang J. Stage-Related Neurotoxicity of BPA in the Development of Zebrafish Embryos. TOXICS 2023; 11:toxics11020177. [PMID: 36851052 PMCID: PMC9963847 DOI: 10.3390/toxics11020177] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 06/10/2023]
Abstract
Bisphenol A (BPA) is one of the most widely produced chemicals in the world used in the production of epoxy resins and polycarbonate plastics. BPA is easily migrated from the outer packaging to the contents. Due to the lipophilic property, BPA is easily accumulated in organisms. Perinatal low-dose BPA exposure alters brain neural development in later generations. In this study, after BPA treatment, the spontaneous movement of zebrafish larvae from the cleavage period to the segmentation period (1-24 hpf) was significantly decreased, with speed decreasing by 18.97% and distance decreasing between 18.4 and 29.7% compared to controls. Transcriptomics analysis showed that 131 genes were significantly differentially expressed in the exposed group during the 1-24 hpf period, among which 39 genes were significantly upregulated and 92 genes were significantly downregulated. The GO enrichment analysis, gene function analysis and real-time quantitative PCR of differentially expressed genes showed that the mRNA level of guanine deaminase (cypin) decreased significantly in the 1-24 hpf period. Moreover, during the 1-24 hpf period, BPA exposure reduced guanine deaminase activity. Therefore, we confirmed that cypin is a key sensitive gene for BPA during this period. Finally, the cypin mRNA microinjection verified that the cypin level of zebrafish larvae was restored, leading to the restoration of the locomotor activity. Taken together, the current results show that the sensitive period of BPA to zebrafish embryos is from the cleavage period to the segmentation period (1-24 hpf), and cypin is a potential target for BPA-induced neurodevelopmental toxicity. This study provides a potential sensitive period and a potential target for the deep understanding of neurodevelopmental toxicity mechanisms caused by BPA.
Collapse
|
8
|
Bagnall-Moreau C, Spielman B, Brimberg L. Maternal brain reactive antibodies profile in autism spectrum disorder: an update. Transl Psychiatry 2023; 13:37. [PMID: 36737600 PMCID: PMC9898547 DOI: 10.1038/s41398-023-02335-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder with multifactorial etiologies involving both genetic and environmental factors. In the past two decades it has become clear that in utero exposure to toxins, inflammation, microbiome, and antibodies (Abs), may play a role in the etiology of ASD. Maternal brain-reactive Abs, present in 10-20% of mothers of a child with ASD, pose a potential risk to the developing brain because they can gain access to the brain during gestation, altering brain development during a critical period. Different maternal anti-brain Abs have been associated with ASD and have been suggested to bind extracellular or intracellular neuronal antigens. Clinical data from various cohorts support the increase in prevalence of such maternal brain-reactive Abs in mothers of a child with ASD compared to mothers of a typically developing child. Animal models of both non-human primates and rodents have provided compelling evidence supporting a pathogenic role of these Abs. In this review we summarize the data from clinical and animal models addressing the role of pathogenic maternal Abs in ASD. We propose that maternal brain-reactive Abs are an overlooked and promising field of research, representing a modifiable risk factor that may account for up to 20% of cases of ASD. More studies are needed to better characterize the Abs that contribute to the risk of having a child with ASD, to understand whether we can we predict such cases of ASD, and to better pinpoint the antigenic specificity of these Abs and their mechanisms of pathogenicity.
Collapse
Affiliation(s)
- Ciara Bagnall-Moreau
- grid.250903.d0000 0000 9566 0634Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, New York, NY USA
| | - Benjamin Spielman
- grid.250903.d0000 0000 9566 0634Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, New York, NY USA ,grid.512756.20000 0004 0370 4759Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY USA
| | - Lior Brimberg
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, New York, NY, USA. .,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
9
|
Kim NH, Lee AY. Growth Factors Upregulated by Uric Acid Affect Guanine Deaminase-Induced Melanogenesis. Biomol Ther (Seoul) 2023; 31:89-96. [PMID: 36549672 PMCID: PMC9810452 DOI: 10.4062/biomolther.2022.137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 12/24/2022] Open
Abstract
Uric acid produced by guanine deaminase (GDA) is involved in photoaging and hyperpigmentation. Reactive oxygen species (ROS) generated by uric acid plays a role in photoaging. However, the mechanism by which uric acid stimulates melanogenesis in GDA-overexpressing keratinocytes is unclear. Keratinocyte-derived paracrine factors have been identified as important mechanisms of ultraviolet-induced melanogenesis. Therefore, the role of paracrine melanogenic growth factors in GDA-induced hypermelanosis mediated by uric acid was examined. The relationships between ROS and these growth factors were examined. Primary cultured normal keratinocytes overexpressed with wild type or mutant GDA and those treated with xanthine or uric acid in the presence or absence of allopurinol, H2O2, or N-acetylcysteine (NAC) were used in this study. Intracellular and extracellular bFGF and SCF levels were increased in keratinocytes by wild type, but not by loss-of-function mutants of GDA overexpression. Culture supernatants from GDA-overexpressing keratinocytes stimulated melanogenesis, which was restored by anti-bFGF and anti-SCF antibodies. Allopurinol treatment reduced the expression levels of bFGF and SCF in both GDA-overexpressing and normal keratinocytes exposed to exogenous xanthine; the exogenous uric acid increased their expression levels. H2O2-stimulated tyrosinase expression and melanogenesis were restored by NAC pretreatment. However, H2O2 or NAC did not upregulate or downregulate bFGF or SCF, respectively. Overall, uric acid could be involved in melanogenesis induced by GDA overexpression in keratinocytes via bFGF and SCF upregulation not via ROS generation.
Collapse
Affiliation(s)
- Nan-Hyung Kim
- Department of Dermatology, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea
| | - Ai-Young Lee
- Department of Dermatology, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea,Corresponding Author E-mail: , Tel: +82-31-961-7250, Fax: +82-31-961-7695
| |
Collapse
|
10
|
Sweet ES, Lange KR, Fenner MR, Tseng CY, Akum BF, Firestein BL. Cypin binds to tubulin heterodimers and microtubule protofilaments and regulates microtubule spacing in developing hippocampal neurons. Mol Cell Neurosci 2022; 123:103783. [PMID: 36208859 DOI: 10.1016/j.mcn.2022.103783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 09/20/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022] Open
Abstract
Cytosolic PSD-95 interactor (cypin) is a multifunctional, guanine deaminase that plays a major role in shaping the morphology of the dendritic arbor of hippocampal and cortical neurons. Cypin catalyzes the Zn2+-dependent deamination of guanine to xanthine, which is then metabolized to uric acid by xanthine oxidase. Cypin binds to tubulin heterodimers via its carboxyl terminal region (amino acids (aa) 350-454), which contains a collapsin response mediator protein (CRMP) homology domain (aa 350-403). Moreover, this region alone is not sufficient to facilitate microtubule polymerization; therefore, additional cypin regions must be involved in this process. Here, we asked whether cypin binds to fully formed microtubules and how overexpression of cypin regulates the microtubule cytoskeleton in dendrites of cultured hippocampal neurons. Protein-protein docking strategies confirm that the cypin homodimer binds to tubulin heterodimers via amino acids within aa 350-454. Biochemical pull-down data suggest that aa 1-220 are necessary for cypin binding to soluble tubulin heterodimers and to taxol-stabilized microtubules. Molecular docking of the cypin homodimer to soluble tubulin heterodimers reveals a consistently observed docking pose using aa 47-71, 113-118, 174-178, and 411-418, which is consistent with our biochemical data. Additionally, overexpression of cypin in hippocampal neurons results in decreased spacing between microtubules. Our results suggest that several protein domains facilitate cypin-mediated polymerization of tubulin heterodimers into microtubules, possibly through a mechanism whereby cypin dimers bind to multiple tubulin heterodimers.
Collapse
Affiliation(s)
- Eric S Sweet
- Department of Biology, West Chester University, West Chester, PA, United States of America
| | - Keith R Lange
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America; Molecular Biosciences Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America
| | - Madeleine R Fenner
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America
| | - Chia-Yi Tseng
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America; Neurosciences Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America
| | - Barbara F Akum
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America; Molecular Biosciences Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America.
| |
Collapse
|
11
|
Angkustsiri K, Fussell JJ, Bennett A, Schauer J, Ramirez-Celis A, Hansen RL, Van de Water J. Pilot Study of Maternal Autoantibody-Related Autism. J Dev Behav Pediatr 2022; 43:465-471. [PMID: 35943360 PMCID: PMC9561005 DOI: 10.1097/dbp.0000000000001100] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 04/06/2022] [Indexed: 02/04/2023]
Abstract
OBJECTIVE The objective of this study was to investigate the presence of maternal autoantibody-related autism spectrum disorder (MAR-ASD) in 2 geographically distinct DBPNet clinical sites (Pennsylvania and Arkansas). MAR-ASD is a biologically defined subtype of ASD that is defined by the presence of autoantibodies specific to proteins in the fetal brain and present in approximately 20% of a Northern California sample but has not been studied in other states. METHODS Sixty-eight mothers of children with ASD were recruited from 2 DBPNet clinics and provided blood samples. Mothers also completed behavioral questionnaires about their children, and data from the child's clinical diagnostic assessment were abstracted. RESULTS The mean age of mothers was 38.5 ± 6.1 years, and the mean age of children was 8.3 ± 2.7 years. MAR-ASD was present in 24% of the sample and similar across sites. Children of +MAR mothers had more severe autism symptoms as measured by Autism Diagnostic Observation Schedule comparison scores (W = 3604; p < 0.001) and the Social Communication Questionnaire (W = 4556; p < 0.001). There were no differences in IQ, adaptive function, or aberrant behavior. CONCLUSION MAR-ASD is a subtype of autism that is present in similar frequencies across 3 states and related to autism severity.
Collapse
Affiliation(s)
- Kathleen Angkustsiri
- Developmental Behavioral Pediatrics, Department of
Pediatrics, University of California Davis Health, Sacramento, CA
- UC Davis MIND Institute, Sacramento, California
| | - Jill J. Fussell
- Developmental-Behavioral Pediatrics and Rehabilitative
Medicine, Department of Pediatrics, University of Arkansas for Medical Sciences,
Little Rock, AR, USA
| | - Amanda Bennett
- Developmental and Behavioral Pediatrics, Department of
Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Joseph Schauer
- Rheumatology, Allergy, and Clinical Immunology, Department
of Internal Medicine, University of California, Davis, CA
| | - Alexandra Ramirez-Celis
- Rheumatology, Allergy, and Clinical Immunology, Department
of Internal Medicine, University of California, Davis, CA
| | - Robin L. Hansen
- Developmental Behavioral Pediatrics, Department of
Pediatrics, University of California Davis Health, Sacramento, CA
- UC Davis MIND Institute, Sacramento, California
| | - Judy Van de Water
- UC Davis MIND Institute, Sacramento, California
- Rheumatology, Allergy, and Clinical Immunology, Department
of Internal Medicine, University of California, Davis, CA
| |
Collapse
|
12
|
Ramirez-Celis A, Croen LA, Yoshida CK, Alexeeff SE, Schauer J, Yolken RH, Ashwood P, Van de Water J. Maternal autoantibody profiles as biomarkers for ASD and ASD with co-occurring intellectual disability. Mol Psychiatry 2022; 27:3760-3767. [PMID: 35618885 PMCID: PMC9708563 DOI: 10.1038/s41380-022-01633-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/05/2022] [Accepted: 05/12/2022] [Indexed: 02/08/2023]
Abstract
Maternal autoantibody-related ASD (MAR ASD) is a subtype of autism in which pathogenic maternal autoantibodies (IgG) cross the placenta, access the developing brain, and cause neurodevelopmental alterations and behaviors associated with autism in the exposed offspring. We previously reported maternal IgG response to eight proteins (CRMP1, CRMP2, GDA LDHA, LDHB, NSE, STIP1, and YBOX) and that reactivity to nine specific combinations of these proteins (MAR ASD patterns) was predictive of ASD risk. The aim of the current study was to validate the previously identified MAR ASD patterns (CRMP1 + GDA, CRMP1 + CRMP2, NSE + STIP1, CRMP2 + STIP1, LDHA + YBOX, LDHB + YBOX, GDA + YBOX, STIP1 + YBOX, and CRMP1 + STIP1) and their accuracy in predicting ASD risk in a prospective cohort employing maternal samples collected prior to parturition. We used prenatal plasma from mothers of autistic children with or without co-occurring intellectual disability (ASD = 540), intellectual disability without autism (ID = 184) and general population controls (GP = 420) collected by the Early Markers for Autism (EMA) study. We found reactivity to one or more of the nine previously identified MAR ASD patterns in 10% of the ASD group compared with 4% of the ID group and 1% of the GP controls (ASD vs GP: Odds Ratio (OR) = 7.81, 95% Confidence Interval (CI) 3.32 to 22.43; ASD vs ID: OR = 2.77, 95% CI (1.19-7.47)) demonstrating that the MAR ASD patterns are strongly associated with the ASD group and could be used to assess ASD risk prior to symptom onset. The pattern most strongly associated with ASD was CRMP1 + CRMP2 and increased the odds for an ASD diagnosis 16-fold (3.32 to >999.99). In addition, we found that several of these specific MAR ASD patterns were strongly associated with ASD with intellectual disability (ASD + ID) and others associated with ASD without ID (ASD-no ID). Prenatal screening for these MAR patterns may lead to earlier identification of ASD and facilitate access to the appropriate early intervention services based on each child's needs.
Collapse
Affiliation(s)
- Alexandra Ramirez-Celis
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, One Shields Avenue, University of California, Davis, CA, 95616, USA
| | - Lisa A Croen
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, 94612, USA
| | - Cathleen K Yoshida
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, 94612, USA
| | - Stacey E Alexeeff
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, 94612, USA
| | - Joseph Schauer
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, One Shields Avenue, University of California, Davis, CA, 95616, USA
| | - Robert H Yolken
- Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul Ashwood
- UC Davis MIND Institute, 2825 50th St, Sacramento, CA, 95817, USA
- Department of Medical Microbiology and Immunology, One Shields Avenue, University of California, Davis, CA, 95616, USA
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, One Shields Avenue, University of California, Davis, CA, 95616, USA.
- UC Davis MIND Institute, 2825 50th St, Sacramento, CA, 95817, USA.
| |
Collapse
|
13
|
Yang J, Zeng J, Lu J. Mechanisms of ultraviolet-induced melasma formation: A review. J Dermatol 2022; 49:1201-1210. [PMID: 35946331 DOI: 10.1111/1346-8138.16542] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 07/03/2022] [Accepted: 07/26/2022] [Indexed: 11/30/2022]
Abstract
Melasma, a pigmentation disorder, commonly occurs in exposed skin areas and can be attributed to several factors. Ultraviolet radiation (UVR) is the primary factor that induces and aggravates melasma. Considering gene expression, exposed skin areas experience abnormal gene expression, involving melanin metabolism, oxidative stress, impaired skin barrier function, and abnormal composition of nerve factors. From a histological perspective, UVR can cause basement membrane collapse, melanocyte sinking, and disorders of skin lipid metabolism. Emerging therapies have focused on these pathological alterations in melasma, including platelet-rich plasma, mesotherapy, and phytochemicals. Understanding the role of UVR in the development of melasma can facilitate early prevention and highlight the future direction of melasma treatment.
Collapse
Affiliation(s)
- Jian Yang
- The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jinrong Zeng
- The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jianyun Lu
- The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
14
|
Liang C, Carrel D, Singh NK, Hiester LL, Fanget I, Kim H, Firestein BL. Carboxypeptidase E Independently Changes Microtubule Glutamylation, Dendritic Branching, and Neuronal Migration. ASN Neuro 2022; 14:17590914211062765. [PMID: 35014548 PMCID: PMC8755936 DOI: 10.1177/17590914211062765] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Neuronal migration and dendritogenesis are dependent on dynamic changes to the microtubule (MT) network. Among various factors that regulate MT dynamics and stability, post-translational modifications (PTMs) of MTs play a critical role in conferring specificity of regulatory protein binding to MTs. Thus, it is important to understand the regulation of PTMs during brain development as multiple developmental processes are dependent on MTs. In this study, we identified that carboxypeptidase E (CPE) changes tubulin polyglutamylation, a major PTM in the brain, and we examine the impact of CPE-mediated changes to polyglutamylation on cortical neuron migration and dendrite morphology. We show, for the first time, that overexpression of CPE increases the level of polyglutamylated α-tubulin while knockdown decreases the level of polyglutamylation. We also demonstrate that CPE-mediated changes to polyglutamylation are dependent on the CPE zinc-binding motif and that this motif is necessary for CPE action on p150Glued localization. However, overexpression of a CPE mutant that does not increase MT glutamylation mimics the effects of overexpression of wild type CPE on dendrite branching. Furthermore, although overexpression of wild type CPE does not alter cortical neuron migration, overexpression of the mutant may act in a dominant-negative manner as it decreases the number of neurons that reach the cortical plate (CP), as we previously reported for CPE knockdown. Overall, our data suggest that CPE changes MT glutamylation and redistribution of p150Glued and that this function of CPE is independent of its role in shaping dendrite development but plays a partial role in regulating cortical neuron migration.
Collapse
Affiliation(s)
- Chen Liang
- Department of Cell Biology and Neuroscience, Rutgers, 242612The State University of New Jersey, Piscataway, NJ, USA.,Molecular Biosciences Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Damien Carrel
- SPPIN Laboratory, 555089Université de Paris, Centre National de la Recherche Scientifique UMR 8003, Paris, France
| | - Nisha K Singh
- Department of Cell Biology and Neuroscience, Rutgers, 242612The State University of New Jersey, Piscataway, NJ, USA.,Molecular Biosciences Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Liam L Hiester
- Department of Cell Biology and Neuroscience, Rutgers, 242612The State University of New Jersey, Piscataway, NJ, USA
| | - Isabelle Fanget
- SPPIN Laboratory, 555089Université de Paris, Centre National de la Recherche Scientifique UMR 8003, Paris, France
| | - Hyuck Kim
- Department of Cell Biology and Neuroscience, Rutgers, 242612The State University of New Jersey, Piscataway, NJ, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, 242612The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
15
|
IRS1 expression in hippocampus is age-dependent and is required for mature spine maintenance and neuritogenesis. Mol Cell Neurosci 2021; 118:103693. [PMID: 34942345 DOI: 10.1016/j.mcn.2021.103693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 11/21/2022] Open
Abstract
Insulin and insulin-like growth factor type I (IGF-1) play prominent roles in brain activity throughout the lifespan. Insulin/IGF1 signaling starts with the activation of the intracellular insulin receptor substrates (IRS). In this work, we performed a comparative study of IRS1 and IRS2, together with the IGF1 (IGF1R) and insulin (IR) receptor expression in the hippocampus and prefrontal cortex during development. We found that IRS1 and IRS2 expression is prominent during development and declines in the aged hippocampus, contrary to IR, which increases in adulthood and aging. In contrast, IGF1R expression is unaffected by age. Expression patterns are similar in the prefrontal cortex. Neurite development occurs postnatally in the rodent hippocampus and cortex, and it declines in the mature and aged brain and is influenced by trophic factors. In our previous work, we demonstrated that knockdown of IRS1 by shRNA impairs learning and reduces synaptic plasticity in a rat model, as measured by synaptophysin puncta in axons. In this study, we report that shIRS1 alters spine maturation in adult hilar hippocampal neurons. Lastly, to understand the role of IRS1 in neuronal neurite tree, we transfect shIRS1 into primary neuronal cultures and observed that shIRS1 reduced neurite branching and neurite length. Our results demonstrate that IRS1/2 and insulin/IGF1 receptors display different age-dependent expression profiles and that IRS1 is required for spine maturation, demonstrating a novel role for IRS1 in synaptic plasticity.
Collapse
|
16
|
Gevezova M, Sarafian V, Anderson G, Maes M. Inflammation and Mitochondrial Dysfunction in Autism Spectrum Disorder. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 19:320-333. [PMID: 32600237 DOI: 10.2174/1871527319666200628015039] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/30/2020] [Accepted: 05/01/2020] [Indexed: 12/11/2022]
Abstract
Autism Spectrum Disorders (ASD) is a severe childhood psychiatric condition with an array of cognitive, language and social impairments that can significantly impact family life. ASD is classically characterized by reduced communication skills and social interactions, with limitations imposed by repetitive patterns of behavior, interests, and activities. The pathophysiology of ASD is thought to arise from complex interactions between environmental and genetic factors within the context of individual development. A growing body of research has raised the possibility of identifying the aetiological causes of the disorder. This review highlights the roles of immune-inflammatory pathways, nitro-oxidative stress and mitochondrial dysfunctions in ASD pathogenesis and symptom severity. The role of NK-cells, T helper, T regulatory and B-cells, coupled with increased inflammatory cytokines, lowered levels of immune-regulatory cytokines, and increased autoantibodies and microglial activation is elucidated. It is proposed that alterations in mitochondrial activity and nitrooxidative stress are intimately associated with activated immune-inflammatory pathways. Future research should determine as to whether the mitochondria, immune-inflammatory activity and nitrooxidative stress changes in ASD affect the development of amygdala-frontal cortex interactions. A number of treatment implications may arise, including prevention-orientated prenatal interventions, treatment of pregnant women with vitamin D, and sodium butyrate. Treatments of ASD children and adults with probiotics, sodium butyrate and butyrate-inducing diets, antipurinergic therapy with suramin, melatonin, oxytocin and taurine are also discussed.
Collapse
Affiliation(s)
- Maria Gevezova
- Department of Medical Biology, Faculty of Medicine, Medical University-Plovdiv, Plovdiv, Bulgaria,Research Institute at Medical University-Plovdiv, Plovdiv, Bulgaria
| | - Victoria Sarafian
- Department of Medical Biology, Faculty of Medicine, Medical University-Plovdiv, Plovdiv, Bulgaria,Research Institute at Medical University-Plovdiv, Plovdiv, Bulgaria
| | | | - Michael Maes
- Department of Medical Biology, Faculty of Medicine, Medical University-Plovdiv, Plovdiv, Bulgaria,Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand,IMPACT Strategic Research Center, Deakin University, Geelong, Australia
| |
Collapse
|
17
|
Bucardo MS, Wu Y, Ludford PT, Li Y, Fin A, Tor Y. Real-Time Monitoring of Human Guanine Deaminase Activity by an Emissive Guanine Analog. ACS Chem Biol 2021; 16:1208-1214. [PMID: 34190533 PMCID: PMC9109600 DOI: 10.1021/acschembio.1c00232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Guanine deaminase (GDA) deaminates guanine to xanthine. Despite its significance, the study of human GDA remains limited compared to other metabolic deaminases. As a result, its substrate and inhibitor repertoire are limited, and effective real-time activity, inhibitory, and discovery assays are missing. Herein, we explore two emissive heterocyclic cores, based on thieno[3,4-d]pyrimidine (thN) and isothiazole[4,3-d]pyrimidine (tzN), as surrogate GDA substrates. We demonstrate that, unlike the thieno analog, thGN, the isothiazolo guanine surrogate, tzGN, does undergo effective enzymatic deamination by GDA and yields the spectroscopically distinct xanthine analog, tzXN. Further, we showcase the potential of this fluorescent nucleobase surrogate to provide a visible spectral window for a real-time study of GDA and its inhibition.
Collapse
Affiliation(s)
- Marcela S Bucardo
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - You Wu
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Paul T Ludford
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Yao Li
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Andrea Fin
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Yitzhak Tor
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
18
|
Morcillo P, Cordero H, Ijomone OM, Ayodele A, Bornhorst J, Gunther L, Macaluso FP, Bowman AB, Aschner M. Defective Mitochondrial Dynamics Underlie Manganese-Induced Neurotoxicity. Mol Neurobiol 2021; 58:3270-3289. [PMID: 33666854 PMCID: PMC9009155 DOI: 10.1007/s12035-021-02341-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/23/2021] [Indexed: 12/17/2022]
Abstract
Perturbations in mitochondrial dynamics have been observed in most neurodegenerative diseases. Here, we focus on manganese (Mn)-induced Parkinsonism-like neurodegeneration, a disorder associated with the preferential of Mn in the basal ganglia where the mitochondria are considered an early target. Despite the extensive characterization of the clinical presentation of manganism, the mechanism by which Mn mediated mitochondrial toxicity is unclear. In this study we hypothesized whether Mn exposure alters mitochondrial activity, including axonal transport of mitochondria and mitochondrial dynamics, morphology, and network. Using primary neuron cultures exposed to 100 μM Mn (which is considered the threshold of Mn toxicity in vitro) and intraperitoneal injections of MnCl2 (25mg/kg) in rat, we observed that Mn increased mitochondrial fission mediated by phosphorylation of dynamin-related protein-1 at serine 616 (p-s616-DRP1) and decreased mitochondrial fusion proteins (MFN1 and MFN2) leading to mitochondrial fragmentation, defects in mitochondrial respiratory capacity, and mitochondrial ultrastructural damage in vivo and in vitro. Furthermore, Mn exposure impaired mitochondrial trafficking by decreasing dynactin (DCTN1) and kinesin-1 (KIF5B) motor proteins and increasing destabilization of the cytoskeleton at protein and gene levels. In addition, mitochondrial communication may also be altered by Mn exposure, increasing the length of nanotunnels to reach out distal mitochondria. These findings revealed an unrecognized role of Mn in dysregulation of mitochondrial dynamics providing a potential explanation of early hallmarks of the disorder, as well as a possible common pathway with neurological disorders arising upon chronic Mn exposure.
Collapse
Affiliation(s)
- Patricia Morcillo
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Hector Cordero
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Omamuyovwi M Ijomone
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- The Neuro-Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Akinyemi Ayodele
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Julia Bornhorst
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Leslie Gunther
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Frank P Macaluso
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
19
|
SREBP-1c Deficiency Affects Hippocampal Micromorphometry and Hippocampus-Dependent Memory Ability in Mice. Int J Mol Sci 2021; 22:ijms22116103. [PMID: 34198910 PMCID: PMC8201143 DOI: 10.3390/ijms22116103] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 11/18/2022] Open
Abstract
Changes in structural and functional neuroplasticity have been implicated in various neurological disorders. Sterol regulatory element-binding protein (SREBP)-1c is a critical regulatory molecule of lipid homeostasis in the brain. Recently, our findings have shown the potential involvement of SREBP-1c deficiency in the alteration of novel modulatory molecules in the hippocampus and occurrence of schizophrenia-like behaviors in mice. However, the possible underlying mechanisms, related to neuronal plasticity in the hippocampus, are yet to be elucidated. In this study, we investigated the hippocampus-dependent memory function and neuronal architecture of hippocampal neurons in SREBP-1c knockout (KO) mice. During the passive avoidance test, SREBP-1c KO mice showed memory impairment. Based on Golgi staining, the dendritic complexity, length, and branch points were significantly decreased in the apical cornu ammonis (CA) 1, CA3, and dentate gyrus (DG) subregions of the hippocampi of SREBP-1c KO mice, compared with those of wild-type (WT) mice. Additionally, significant decreases in the dendritic diameters were detected in the CA3 and DG subregions, and spine density was also significantly decreased in the apical CA3 subregion of the hippocampi of KO mice, compared with that of WT mice. Alterations in the proportions of stubby and thin-shaped dendritic spines were observed in the apical subcompartments of CA1 and CA3 in the hippocampi of KO mice. Furthermore, the corresponding differential decreases in the levels of SREBP-1 expression in the hippocampal subregions (particularly, a significant decrease in the level in the CA3) were detected by immunofluorescence. This study suggests that the contributions of SREBP-1c to the structural plasticity of the mouse hippocampus may have underlain the behavioral alterations. These findings offer insights into the critical role of SREBP-1c in hippocampal functioning in mice.
Collapse
|
20
|
Rodriguez AR, Anderson ED, O'Neill KM, McEwan PP, Vigilante NF, Kwon M, Akum BF, Stawicki TM, Meaney DF, Firestein BL. Cytosolic PSD-95 interactor alters functional organization of neural circuits and AMPA receptor signaling independent of PSD-95 binding. Netw Neurosci 2021; 5:166-197. [PMID: 33688611 PMCID: PMC7935033 DOI: 10.1162/netn_a_00173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/26/2020] [Indexed: 11/04/2022] Open
Abstract
Cytosolic PSD-95 interactor (cypin) regulates many aspects of neuronal development and function, ranging from dendritogenesis to synaptic protein localization. While it is known that removal of postsynaptic density protein-95 (PSD-95) from the postsynaptic density decreases synaptic N-methyl-D-aspartate (NMDA) receptors and that cypin overexpression protects neurons from NMDA-induced toxicity, little is known about cypin's role in AMPA receptor clustering and function. Experimental work shows that cypin overexpression decreases PSD-95 levels in synaptosomes and the PSD, decreases PSD-95 clusters/μm2, and increases mEPSC frequency. Analysis of microelectrode array (MEA) data demonstrates that cypin or cypinΔPDZ overexpression increases sensitivity to CNQX (cyanquixaline) and AMPA receptor-mediated decreases in spike waveform properties. Network-level analysis of MEA data reveals that cypinΔPDZ overexpression causes networks to be resilient to CNQX-induced changes in local efficiency. Incorporating these findings into a computational model of a neural circuit demonstrates a role for AMPA receptors in cypin-promoted changes to networks and shows that cypin increases firing rate while changing network functional organization, suggesting cypin overexpression facilitates information relay but modifies how information is encoded among brain regions. Our data show that cypin promotes changes to AMPA receptor signaling independent of PSD-95 binding, shaping neural circuits and output to regions beyond the hippocampus.
Collapse
Affiliation(s)
- Ana R Rodriguez
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Erin D Anderson
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Kate M O'Neill
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Przemyslaw P McEwan
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | | | - Munjin Kwon
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Barbara F Akum
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Tamara M Stawicki
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
21
|
Quach TT, Stratton HJ, Khanna R, Kolattukudy PE, Honnorat J, Meyer K, Duchemin AM. Intellectual disability: dendritic anomalies and emerging genetic perspectives. Acta Neuropathol 2021; 141:139-158. [PMID: 33226471 PMCID: PMC7855540 DOI: 10.1007/s00401-020-02244-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022]
Abstract
Intellectual disability (ID) corresponds to several neurodevelopmental disorders of heterogeneous origin in which cognitive deficits are commonly associated with abnormalities of dendrites and dendritic spines. These histological changes in the brain serve as a proxy for underlying deficits in neuronal network connectivity, mostly a result of genetic factors. Historically, chromosomal abnormalities have been reported by conventional karyotyping, targeted fluorescence in situ hybridization (FISH), and chromosomal microarray analysis. More recently, cytogenomic mapping, whole-exome sequencing, and bioinformatic mining have led to the identification of novel candidate genes, including genes involved in neuritogenesis, dendrite maintenance, and synaptic plasticity. Greater understanding of the roles of these putative ID genes and their functional interactions might boost investigations into determining the plausible link between cellular and behavioral alterations as well as the mechanisms contributing to the cognitive impairment observed in ID. Genetic data combined with histological abnormalities, clinical presentation, and transgenic animal models provide support for the primacy of dysregulation in dendrite structure and function as the basis for the cognitive deficits observed in ID. In this review, we highlight the importance of dendrite pathophysiology in the etiologies of four prototypical ID syndromes, namely Down Syndrome (DS), Rett Syndrome (RTT), Digeorge Syndrome (DGS) and Fragile X Syndrome (FXS). Clinical characteristics of ID have also been reported in individuals with deletions in the long arm of chromosome 10 (the q26.2/q26.3), a region containing the gene for the collapsin response mediator protein 3 (CRMP3), also known as dihydropyrimidinase-related protein-4 (DRP-4, DPYSL4), which is involved in dendritogenesis. Following a discussion of clinical and genetic findings in these syndromes and their preclinical animal models, we lionize CRMP3/DPYSL4 as a novel candidate gene for ID that may be ripe for therapeutic intervention.
Collapse
Affiliation(s)
- Tam T Quach
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
- INSERM U1217/CNRS, UMR5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, Tucson, AZ, 85724, USA
| | | | - Jérome Honnorat
- INSERM U1217/CNRS, UMR5310, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Lyon, France
- SynatAc Team, Institut NeuroMyoGène, Lyon, France
| | - Kathrin Meyer
- The Research Institute of Nationwide Children Hospital, Columbus, OH, 43205, USA
- Department of Pediatric, The Ohio State University, Columbus, OH, 43210, USA
| | - Anne-Marie Duchemin
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
22
|
Ramirez-Celis A, Becker M, Nuño M, Schauer J, Aghaeepour N, Van de Water J. Risk assessment analysis for maternal autoantibody-related autism (MAR-ASD): a subtype of autism. Mol Psychiatry 2021; 26:1551-1560. [PMID: 33483694 PMCID: PMC8159732 DOI: 10.1038/s41380-020-00998-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 11/16/2022]
Abstract
The incidence of autism spectrum disorder (ASD) has been rising, however ASD-risk biomarkers remain lacking. We previously identified the presence of maternal autoantibodies to fetal brain proteins specific to ASD, now termed maternal autoantibody-related (MAR) ASD. The current study aimed to create and validate a serological assay to identify ASD-specific maternal autoantibody patterns of reactivity against eight previously identified proteins (CRMP1, CRMP2, GDA, NSE, LDHA, LDHB, STIP1, and YBOX) that are highly expressed in developing brain, and determine the relationship of these reactivity patterns with ASD outcome severity. We used plasma from mothers of children diagnosed with ASD (n = 450) and from typically developing children (TD, n = 342) to develop an ELISA test for each of the protein antigens. We then determined patterns of reactivity a highly significant association with ASD, and discovered several patterns that were ASD-specific (18% in the training set and 10% in the validation set vs. 0% TD). The three main patterns associated with MAR ASD are CRMP1 + GDA (ASD% = 4.2 vs. TD% = 0, OR 31.04, p = <0.0001), CRMP1 + CRMP2 (ASD% = 3.6 vs. TD% = 0, OR 26.08, p = 0.0005) and NSE + STIP1 (ASD% = 3.1 vs. TD% = 0, OR 22.82, p = 0.0001). Additionally, we found that maternal autoantibody reactivity to CRMP1 significantly increases the odds of a child having a higher Autism Diagnostic Observation Schedule (ADOS) severity score (OR 2.3; 95% CI: 1.358-3.987, p = 0.0021). This is the first report that uses machine learning subgroup discovery to identify with 100% accuracy MAR ASD-specific patterns as potential biomarkers of risk for a subset of up to 18% of ASD cases in this study population.
Collapse
Affiliation(s)
- Alexandra Ramirez-Celis
- grid.27860.3b0000 0004 1936 9684Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, One Shields Avenue, University of California, Davis, CA 95616 USA
| | - Martin Becker
- grid.168010.e0000000419368956Department of Anesthesiology, Pain, and Perioperative Medicine, Stanford University, Palo Alto, CA 94305 USA ,grid.168010.e0000000419368956Department of Pediatrics, Stanford University, Palo Alto, CA 94305 USA ,grid.168010.e0000000419368956Department of Biomedical Data Sciences, Stanford University, Palo Alto, CA 94305 USA
| | - Miriam Nuño
- grid.27860.3b0000 0004 1936 9684Department of Public Health Sciences, Division of Biostatistics, One Shields Avenue, University of California, Davis, CA 95616 USA
| | - Joseph Schauer
- grid.27860.3b0000 0004 1936 9684Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, One Shields Avenue, University of California, Davis, CA 95616 USA
| | - Nima Aghaeepour
- grid.168010.e0000000419368956Department of Anesthesiology, Pain, and Perioperative Medicine, Stanford University, Palo Alto, CA 94305 USA ,grid.168010.e0000000419368956Department of Pediatrics, Stanford University, Palo Alto, CA 94305 USA ,grid.168010.e0000000419368956Department of Biomedical Data Sciences, Stanford University, Palo Alto, CA 94305 USA
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, One Shields Avenue, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
23
|
Eslamloo K, Caballero-Solares A, Inkpen SM, Emam M, Kumar S, Bouniot C, Avendaño-Herrera R, Jakob E, Rise ML. Transcriptomic Profiling of the Adaptive and Innate Immune Responses of Atlantic Salmon to Renibacterium salmoninarum Infection. Front Immunol 2020; 11:567838. [PMID: 33193341 PMCID: PMC7656060 DOI: 10.3389/fimmu.2020.567838] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/07/2020] [Indexed: 01/08/2023] Open
Abstract
Bacterial Kidney Disease (BKD), which is caused by a Gram-positive, intracellular bacterial pathogen (Renibacterium salmoninarum), affects salmonids including Atlantic salmon (Salmo salar). However, the transcriptome response of Atlantic salmon to BKD remained unknown before the current study. We used a 44K salmonid microarray platform to characterise the global gene expression response of Atlantic salmon to BKD. Fish (~54 g) were injected with a dose of R. salmoninarum (H-2 strain, 2 × 108 CFU per fish) or sterile medium (control), and then head kidney samples were collected at 13 days post-infection/injection (dpi). Firstly, infection levels of individuals were determined through quantifying the R. salmoninarum level by RNA-based TaqMan qPCR assays. Thereafter, based on the qPCR results for infection level, fish (n = 5) that showed no (control), higher (H-BKD), or lower (L-BKD) infection level at 13 dpi were subjected to microarray analyses. We identified 6,766 and 7,729 differentially expressed probes in the H-BKD and L-BKD groups, respectively. There were 357 probes responsive to the infection level (H-BKD vs. L-BKD). Several adaptive and innate immune processes were dysregulated in R. salmoninarum-infected Atlantic salmon. Adaptive immune pathways associated with lymphocyte differentiation and activation (e.g., lymphocyte chemotaxis, T-cell activation, and immunoglobulin secretion), as well as antigen-presenting cell functions, were shown to be differentially regulated in response to BKD. The infection level-responsive transcripts were related to several mechanisms such as the JAK-STAT signalling pathway, B-cell differentiation and interleukin-1 responses. Sixty-five microarray-identified transcripts were subjected to qPCR validation, and they showed the same fold-change direction as microarray results. The qPCR-validated transcripts studied herein play putative roles in various immune processes including pathogen recognition (e.g., tlr5), antibacterial activity (e.g., hamp and camp), regulation of immune responses (e.g., tnfrsf11b and socs1), T-/B-cell differentiation (e.g., ccl4, irf1 and ccr5), T-cell functions (e.g., rnf144a, il13ra1b and tnfrsf6b), and antigen-presenting cell functions (e.g., fcgr1). The present study revealed diverse immune mechanisms dysregulated by R. salmoninarum in Atlantic salmon, and enhanced the current understanding of Atlantic salmon response to BKD. The identified biomarker genes can be used for future studies on improving the resistance of Atlantic salmon to BKD.
Collapse
Affiliation(s)
- Khalil Eslamloo
- Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | | | - Sabrina M Inkpen
- Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Mohamed Emam
- Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Surendra Kumar
- Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | | | - Ruben Avendaño-Herrera
- Facultad Ciencias de la Vida, Viña del Mar, and FONDAP Interdisciplinary Center for Aquaculture Research (INCAR), Universidad Andrés Bello, Santiago, Chile
| | - Eva Jakob
- Cargill Innovation Center-Colaco, Calbuco, Chile
| | - Matthew L Rise
- Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
24
|
McSweeney C, Dong F, Chen M, Vitale J, Xu L, Crowley N, Luscher B, Zou D, Mao Y. Full function of exon junction complex factor, Rbm8a, is critical for interneuron development. Transl Psychiatry 2020; 10:379. [PMID: 33154347 PMCID: PMC7644723 DOI: 10.1038/s41398-020-01065-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/16/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
The formation of the nervous system requires a balance between proliferation, differentiation, and migration of neural progenitors (NPs). Mutations in genes regulating development impede neurogenesis and lead to neuropsychiatric diseases, including autism spectrum disorders (ASDs) and schizophrenia (SZ). Recently, mutations in nonsense-mediated mRNA decay genes have been associated with ASDs, intellectual disability (ID), and SZ. Here, we examine the function of a gene in the exon junction complex, Rbm8a, in the cortical development. When Rbm8a is selectively knocked out in neural stem cells, the resulting mice exhibit microcephaly, early postnatal lethality, and altered distribution of excitatory neurons in the neocortex. Moreover, Rbm8a haploinsufficiency in the central nervous system decreases cell proliferation in the ganglionic eminences. Parvalbumin+ and neuropeptide Y+ interneurons in the cortex are significantly reduced, and distribution of interneurons are altered. Consistently, neurons in the cortex of conditional knockout (cKO) mice show a significant decrease in GABA frequency. Transcriptomic analysis revealed differentially expressed genes enriched in telencephalon development and mitosis. To further investigate the role of Rbm8a in interneuron differentiation, conditional KO of Rbm8a in NKX2.1 interneuron progenitor cells reduces progenitor proliferation and alters interneuron distributions. Taken together, these data reveal a critical role of Rbm8a in interneuron development, and establish that perturbation of this gene leads to profound cortical deficits.
Collapse
Affiliation(s)
- Colleen McSweeney
- grid.29857.310000 0001 2097 4281Department of Biology, Pennsylvania State University, University Park, PA 16802 USA
| | - Fengping Dong
- grid.29857.310000 0001 2097 4281Department of Biology, Pennsylvania State University, University Park, PA 16802 USA
| | - Miranda Chen
- grid.29857.310000 0001 2097 4281Department of Biology, Pennsylvania State University, University Park, PA 16802 USA
| | - Jessica Vitale
- grid.29857.310000 0001 2097 4281Department of Biology, Pennsylvania State University, University Park, PA 16802 USA
| | - Li Xu
- grid.29857.310000 0001 2097 4281Department of Biology, Pennsylvania State University, University Park, PA 16802 USA ,grid.268505.c0000 0000 8744 8924Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang China
| | - Nicole Crowley
- grid.29857.310000 0001 2097 4281Department of Biology, Pennsylvania State University, University Park, PA 16802 USA
| | - Bernhard Luscher
- grid.29857.310000 0001 2097 4281Department of Biology, Pennsylvania State University, University Park, PA 16802 USA ,grid.29857.310000 0001 2097 4281Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802 USA
| | - Donghua Zou
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA. .,Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China.
| | - Yingwei Mao
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
25
|
Bhaskar S, Gowda J, Prasanna J, Kumar A. Does altering proteasomal activity and trafficking reduce the arborization mediated specific vulnerability of SNpc dopaminergic neurons of Parkinson's disease? Med Hypotheses 2020; 143:110062. [PMID: 32652429 DOI: 10.1016/j.mehy.2020.110062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/27/2020] [Indexed: 01/15/2023]
Abstract
Parkinson's disease (PD) is a late-onset degenerative neuronal disorder and stands second among the neurological disorders with 1% of the total world population being affected. The disease originates majorly due to compromised function of the dopaminergic (DA) neurons in the Substantia Nigra pars compacta (SNpc), but not the ventral tegmental area (VTA) region of the midbrain. The differential susceptibility for degeneration is majorly attributed to morphological, molecular, and electrophysiological heterogeneity existing in DA neurons of SNpc and VTA. Long-range axonal arborization and a higher number of synapses in SNpc DA neurons make it more vulnerable compared to VTA DA neurons. Studies have shown that a decrease in such axonal arborization places DA neurons at decreased risk in PD. The two well established underlying mechanisms are a) As arborization is an energy-demanding process, increased redistribution of mitochondria to the axonal terminals occurs to satisfy the bioenergetic requirement b) The stabilization of axon-promoting factors at the axonal tip is an essential component for enhancing the arborization process. Interfering with any of these two processes would probably alleviate the degeneration of SNpc DA neurons. To accomplish the decreased stability of arborizing factors and thereby increase the resilience of SNpc DA neurons, we hypothesize the activation of anterograde transport-dependent recruitment of proteasomes to axon terminals as one of the most favorable approaches. Understanding this putative avenue of enhancing proteasomal activity and migration to the axonal tip could provide insight into the progression of neurodegeneration in PD and possibly offer a novel therapeutic strategy.
Collapse
Affiliation(s)
- Smitha Bhaskar
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Allalsandra, Yelahanka, Bengaluru 560065, Karnataka, India
| | - Jeevan Gowda
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Allalsandra, Yelahanka, Bengaluru 560065, Karnataka, India
| | - Jyothi Prasanna
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Allalsandra, Yelahanka, Bengaluru 560065, Karnataka, India
| | - Anujith Kumar
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Allalsandra, Yelahanka, Bengaluru 560065, Karnataka, India.
| |
Collapse
|
26
|
Jung JM, Noh TK, Jo SY, Kim SY, Song Y, Kim YH, Chang SE. Guanine Deaminase in Human Epidermal Keratinocytes Contributes to Skin Pigmentation. Molecules 2020; 25:molecules25112637. [PMID: 32517074 PMCID: PMC7321356 DOI: 10.3390/molecules25112637] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/25/2020] [Accepted: 06/02/2020] [Indexed: 01/06/2023] Open
Abstract
Epidermal keratinocytes are considered as the most important neighboring cells that modify melanogenesis. Our previous study used microarray to show that guanine deaminase (GDA) gene expression is highly increased in melasma lesions. Hence, we investigated the role of GDA in skin pigmentation. We examined GDA expression in post-inflammatory hyperpigmentation (PIH) lesions, diagnosed as Riehl’s melanosis. We further investigated the possible role of keratinocyte-derived GDA in melanogenesis by quantitative PCR, immunofluorescence staining, small interfering RNA-based GDA knockdown, and adenovirus-mediated GDA overexpression. We found higher GDA positivity in the hyperpigmentary lesional epidermis than in the perilesional epidermis. Both UVB irradiation and stem cell factor (SCF) plus endothelin-1 (ET-1) were used, which are well-known melanogenic stimuli upregulating GDA expression in both keratinocyte culture alone and keratinocyte and melanocyte coculture. GDA knockdown downregulated melanin content, while GDA overexpression promoted melanogenesis in the coculture. When melanocytes were treated with UVB-exposed keratinocyte-conditioned media, the melanin content was increased. Also, GDA knockdown lowered SCF and ET-1 expression levels in keratinocytes. GDA in epidermal keratinocytes may promote melanogenesis by upregulating SCF and ET-1, suggesting its role in skin hyperpigmentary disorders.
Collapse
Affiliation(s)
- Joon Min Jung
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea; (J.M.J.); (T.K.N.); (S.Y.J.); (S.Y.K.)
| | - Tai Kyung Noh
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea; (J.M.J.); (T.K.N.); (S.Y.J.); (S.Y.K.)
| | - Soo Youn Jo
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea; (J.M.J.); (T.K.N.); (S.Y.J.); (S.Y.K.)
| | - Su Yeon Kim
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea; (J.M.J.); (T.K.N.); (S.Y.J.); (S.Y.K.)
| | - Youngsup Song
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea;
| | - Young-Hoon Kim
- Department of Pharmacology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea
- Correspondence: (Y.-H.K.); (S.E.C.); Tel.: +82-2-3010-4298 (Y.-H.K.); +82-2-3010-3460 (S.E.C.); Fax: +82-2-3010-2941 (Y.-H.K.); +82-2-486-7831 (S.E.C.)
| | - Sung Eun Chang
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea; (J.M.J.); (T.K.N.); (S.Y.J.); (S.Y.K.)
- Correspondence: (Y.-H.K.); (S.E.C.); Tel.: +82-2-3010-4298 (Y.-H.K.); +82-2-3010-3460 (S.E.C.); Fax: +82-2-3010-2941 (Y.-H.K.); +82-2-486-7831 (S.E.C.)
| |
Collapse
|
27
|
Omelchenko A, Menon H, Donofrio SG, Kumar G, Chapman HM, Roshal J, Martinez-Montes ER, Wang TL, Spaller MR, Firestein BL. Interaction Between CRIPT and PSD-95 Is Required for Proper Dendritic Arborization in Hippocampal Neurons. Mol Neurobiol 2020; 57:2479-2493. [PMID: 32157575 PMCID: PMC7176523 DOI: 10.1007/s12035-020-01895-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 02/24/2020] [Indexed: 01/10/2023]
Abstract
CRIPT, the cysteine-rich PDZ-binding protein, binds to the third PDZ domain of PSD-95 (postsynaptic density protein 95) family proteins and directly binds microtubules, linking PSD-95 family proteins to the neuronal cytoskeleton. Here, we show that overexpression of a full-length CRIPT leads to a modest decrease, and knockdown of CRIPT leads to an increase in dendritic branching in cultured rat hippocampal neurons. Overexpression of truncated CRIPT lacking the PDZ domain-binding motif, which does not bind to PSD-95, significantly decreases dendritic arborization. Conversely, overexpression of a full-length CRIPT significantly increases the number of immature and mature dendritic spines, and this effect is not observed when CRIPT∆PDZ is overexpressed. Competitive inhibition of CRIPT binding to the third PDZ domain of PSD-95 with PDZ3-binding peptides resulted in differential effects on dendritic arborization based on the origin of respective peptide sequence. These results highlight multifunctional roles of CRIPT during development and underscore the significance of the interaction between CRIPT and the third PDZ domain of PSD-95.
Collapse
Affiliation(s)
- Anton Omelchenko
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
- Neuroscience Graduate Program, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Harita Menon
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Sarah G Donofrio
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Gaurav Kumar
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Heidi M Chapman
- Geisel School of Medicine, Department of Medical Education and Norris Cotton Cancer Center, Dartmouth College, Lebanon, NH, 03756, USA
| | - Joshua Roshal
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Eduardo R Martinez-Montes
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Tiffany L Wang
- Geisel School of Medicine, Department of Medical Education and Norris Cotton Cancer Center, Dartmouth College, Lebanon, NH, 03756, USA
| | - Mark R Spaller
- Geisel School of Medicine, Department of Medical Education and Norris Cotton Cancer Center, Dartmouth College, Lebanon, NH, 03756, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854-8082, USA.
| |
Collapse
|
28
|
Kadoyama K, Matsuura K, Takano M, Otani M, Tomiyama T, Mori H, Matsuyama S. Proteomic analysis involved with synaptic plasticity improvement by GABA A receptor blockade in hippocampus of a mouse model of Alzheimer's disease. Neurosci Res 2020; 165:61-68. [PMID: 32348793 DOI: 10.1016/j.neures.2020.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 04/03/2020] [Accepted: 04/15/2020] [Indexed: 12/31/2022]
Abstract
GABAergic system plays a part in synaptic plasticity in the hippocampus. We had reported a long-term potentiation (LTP)-like facilitation in vivo, known as synaptic plasticity, through GABAA receptor blockade by bicuculline and the expression of proteins involved with this synaptic plasticity in mouse hippocampus. In the present study, we aimed to show improvement of impaired synaptic plasticity through GABAA receptor blockade and to clarify the molecular mechanisms involved with this improvement in the hippocampus of mice overexpressing human amyloid precursor protein with the E693Δ mutation (APPOSK-Tg) as an Alzheimer's disease model showing impaired synaptic plasticity. Electrophysiological study showed that the LTP-like facilitation expressed with application of bicuculline in vivo was significantly greater than impaired tetanic LTP in APPOSK-Tg mice, which was improved by bicuculline. Proteomic analysis showed that the expression of 11 proteins in the hippocampus was significantly changed 8 h after bicuculline application to APPOSK-Tg mice. The identified proteins could be functionally classified as chaperone, cytoskeletal protein, energy metabolism, metabolism, neuronal development, and synaptic component. Additionally, western blotting validated the changes in four proteins. We therefore propose that the improvement of impaired synaptic plasticity through GABAA receptor blockade could be mediated by the changed expression of these proteins.
Collapse
Affiliation(s)
- Keiichi Kadoyama
- Department of Pharmaceutical Health Care, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, Himeji 670-8524, Japan
| | - Kenji Matsuura
- Faculty of Pharmacy, Osaka-Ohtani University, Tondabayashi 584-8540, Japan
| | - Masaoki Takano
- Department of Life Sciences Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe 650-8586, Japan
| | - Mieko Otani
- Department of Life Sciences Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe 650-8586, Japan
| | - Takami Tomiyama
- Department of Translational Neuroscience, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Hiroshi Mori
- Department of Clinical Neuroscience, Osaka City University Medical School, Osaka 545-8585, Japan
| | - Shogo Matsuyama
- Biosignal Research Center, Kobe University, Kobe 657-8501, Japan.
| |
Collapse
|
29
|
Rodríguez AR, O'Neill KM, Swiatkowski P, Patel MV, Firestein BL. Overexpression of cypin alters dendrite morphology, single neuron activity, and network properties via distinct mechanisms. J Neural Eng 2019; 15:016020. [PMID: 29091046 DOI: 10.1088/1741-2552/aa976a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE This study investigates the effect that overexpression of cytosolic PSD-95 interactor (cypin), a regulator of synaptic PSD-95 protein localization and a core regulator of dendrite branching, exerts on the electrical activity of rat hippocampal neurons and networks. APPROACH We cultured rat hippocampal neurons and used lipid-mediated transfection and lentiviral gene transfer to achieve high levels of cypin or cypin mutant (cypinΔPDZ; PSD-95 non-binding) expression cellularly and network-wide, respectively. MAIN RESULTS Our analysis revealed that although overexpression of cypin and cypinΔPDZ increase dendrite numbers and decrease spine density, cypin and cypinΔPDZ distinctly regulate neuronal activity. At the single cell level, cypin promotes decreases in bursting activity while cypinΔPDZ reduces sEPSC frequency and further decreases bursting compared to cypin. At the network level, by using the Fano factor as a measure of spike count variability, cypin overexpression results in an increase in variability of spike count, and this effect is abolished when cypin cannot bind PSD-95. This variability is also dependent on baseline activity levels and on mean spike rate over time. Finally, our spike sorting data show that overexpression of cypin results in a more complex distribution of spike waveforms and that binding to PSD-95 is essential for this complexity. SIGNIFICANCE Our data suggest that dendrite morphology does not play a major role in cypin action on electrical activity.
Collapse
Affiliation(s)
- Ana R Rodríguez
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, United States of America. Graduate Program in Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, United States of America
| | | | | | | | | |
Collapse
|
30
|
Mazón-Cabrera R, Vandormael P, Somers V. Antigenic Targets of Patient and Maternal Autoantibodies in Autism Spectrum Disorder. Front Immunol 2019; 10:1474. [PMID: 31379804 PMCID: PMC6659315 DOI: 10.3389/fimmu.2019.01474] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/13/2019] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder whose behavioral symptoms become apparent in early childhood. The underlying pathophysiological mechanisms are only partially understood and the clinical manifestations are heterogeneous in nature, which poses a major challenge for diagnosis, prognosis and intervention. In the last years, an important role of a dysregulated immune system in ASD has emerged, but the mechanisms connecting this to a disruption of brain development are still largely unknown. Although ASD is not considered as a typical autoimmune disease, self-reactive antibodies or autoantibodies against a wide variety of targets have been found in a subset of ASD patients. In addition, autoantibodies reactive to fetal brain proteins have also been described in the prenatal stage of neurodevelopment, where they can be transferred from the mother to the fetus by transplacental transport. In this review, we give an extensive overview of the antibodies described in ASD according to their target antigens, their different origins, and timing of exposure during neurodevelopment.
Collapse
Affiliation(s)
| | | | - Veerle Somers
- Biomedical Research Institute, Faculty of Medicine and Life Science, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
31
|
Patel MV, Sewell E, Dickson S, Kim H, Meaney DF, Firestein BL. A Role for Postsynaptic Density 95 and Its Binding Partners in Models of Traumatic Brain Injury. J Neurotrauma 2019; 36:2129-2138. [DOI: 10.1089/neu.2018.6291] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Mihir V. Patel
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
- Graduate Program in Neurosciences, Rutgers University, Piscataway, New Jersey
| | - Emily Sewell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Samantha Dickson
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hyuck Kim
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| | - David F. Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bonnie L. Firestein
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
32
|
Slater PG, Cammarata GM, Monahan C, Bowers JT, Yan O, Lee S, Lowery LA. Characterization of Xenopus laevis guanine deaminase reveals new insights for its expression and function in the embryonic kidney. Dev Dyn 2019; 248:296-305. [PMID: 30682232 DOI: 10.1002/dvdy.14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/18/2018] [Accepted: 01/21/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The mammalian guanine deaminase (GDA), called cypin, is important for proper neural development, by regulating dendritic arborization through modulation of microtubule (MT) dynamics. Additionally, cypin can promote MT assembly in vitro. However, it has never been tested whether cypin (or other GDA orthologs) binds to MTs or modulates MT dynamics. Here, we address these questions and characterize Xenopus laevis GDA (Gda) for the first time during embryonic development. RESULTS We find that exogenously expressed human cypin and Gda both display a cytosolic distribution in primary embryonic cells. Furthermore, while expression of human cypin can promote MT polymerization, Xenopus Gda has no effect. Additionally, we find that the tubulin-binding collapsin response mediator protein (CRMP) homology domain is only partially conserved between cypin and Gda. This likely explains the divergence in function, as we discovered that the cypin region containing the CRMP homology and PDZ-binding domain is necessary for regulating MT dynamics. Finally, we observed that gda is strongly expressed in the kidneys during late embryonic development, although it does not appear to be critical for kidney development. CONCLUSIONS Together, these results suggest that GDA has diverged in function between mammals and amphibians, and that mammalian GDA plays an indirect role in regulating MT dynamics. Developmental Dynamics 248:296-305, 2019. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Paula G Slater
- Boston College, Department of Biology, Chestnut Hill, Massachusetts
| | | | - Connor Monahan
- Boston College, Department of Biology, Chestnut Hill, Massachusetts
| | - Jackson T Bowers
- Boston College, Department of Biology, Chestnut Hill, Massachusetts
| | - Oliver Yan
- Boston College, Department of Biology, Chestnut Hill, Massachusetts
| | - Sangmook Lee
- Boston College, Department of Biology, Chestnut Hill, Massachusetts
| | | |
Collapse
|
33
|
Jones KL, Van de Water J. Maternal autoantibody related autism: mechanisms and pathways. Mol Psychiatry 2019; 24:252-265. [PMID: 29934547 PMCID: PMC6784837 DOI: 10.1038/s41380-018-0099-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 04/12/2018] [Accepted: 04/23/2018] [Indexed: 12/16/2022]
Abstract
It has been estimated that autism spectrum disorder (ASD) now affects 1 in 59 children in the United States. Although the cause(s) of ASD remain largely unknown, it is becoming increasingly apparent that ASD can no longer be defined simply as a behavioral disorder, but is in effect a rather complex and highly heterogeneous biological disorder. Up until recently the brain was thought to be "immune privileged." However, it is now known that the immune system plays critical roles in the development and functioning of the brain throughout life. Recent evidence from multiple investigators has illustrated the deleterious role that dysregulation of the maternal immune system during gestation can play in the manifestation of changes in neurodevelopment, resulting in the development of neurobehavioral disorders such as ASD. One potential etiologic pathway through which the maternal immune system can interfere with neurodevelopment is through maternal autoantibodies that recognize proteins in the developing fetal brain. This mechanism of pathogenesis is now thought to lead to a subphenotype of ASD that has been termed maternal autoantibody related (MAR) ASD. This review provides an overview of the current research implicating the presence of brain-reactive maternal autoantibodies as a risk factor for MAR ASD.
Collapse
Affiliation(s)
- Karen L. Jones
- Rheumatology/Allergy and Clinical Immunology, University of California, 451 E. Health Sciences Drive, Suite 6510 GBSF, Davis, CA 95616, USA,The M.I.N.D. Institute, University of California, Davis, CA 95616, USA
| | - Judy Van de Water
- Rheumatology/Allergy and Clinical Immunology, University of California, 451 E. Health Sciences Drive, Suite 6510 GBSF, Davis, CA, 95616, USA. .,The M.I.N.D. Institute, University of California, Davis, CA, 95616, USA. .,NIEHS Center for Children's Environmental Health, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
34
|
Swiatkowski P, Sewell E, Sweet ES, Dickson S, Swanson RA, McEwan SA, Cuccolo N, McDonnell ME, Patel MV, Varghese N, Morrison B, Reitz AB, Meaney DF, Firestein BL. Cypin: A novel target for traumatic brain injury. Neurobiol Dis 2018; 119:13-25. [PMID: 30031156 DOI: 10.1016/j.nbd.2018.07.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/06/2018] [Accepted: 07/17/2018] [Indexed: 12/13/2022] Open
Abstract
Cytosolic PSD-95 interactor (cypin), the primary guanine deaminase in the brain, plays key roles in shaping neuronal circuits and regulating neuronal survival. Despite this pervasive role in neuronal function, the ability for cypin activity to affect recovery from acute brain injury is unknown. A key barrier in identifying the role of cypin in neurological recovery is the absence of pharmacological tools to manipulate cypin activity in vivo. Here, we use a small molecule screen to identify two activators and one inhibitor of cypin's guanine deaminase activity. The primary screen identified compounds that change the initial rate of guanine deamination using a colorimetric assay, and secondary screens included the ability of the compounds to protect neurons from NMDA-induced injury and NMDA-induced decreases in frequency and amplitude of miniature excitatory postsynaptic currents. Hippocampal neurons pretreated with activators preserved electrophysiological function and survival after NMDA-induced injury in vitro, while pretreatment with the inhibitor did not. The effects of the activators were abolished when cypin was knocked down. Administering either cypin activator directly into the brain one hour after traumatic brain injury significantly reduced fear conditioning deficits 5 days after injury, while delivering the cypin inhibitor did not improve outcome after TBI. Together, these data demonstrate that cypin activation is a novel approach for improving outcome after TBI and may provide a new pathway for reducing the deficits associated with TBI in patients.
Collapse
Affiliation(s)
- Przemyslaw Swiatkowski
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA; Graduate Program in Molecular Biosciences, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Emily Sewell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104-6391, USA
| | - Eric S Sweet
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA; Graduate Program in Neurosciences, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Samantha Dickson
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104-6391, USA
| | - Rachel A Swanson
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Sara A McEwan
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA; Graduate Program in Neurosciences, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Nicholas Cuccolo
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Mark E McDonnell
- Fox Chase Chemical Diversity Center, Inc., Doylestown, PA 18902, USA
| | - Mihir V Patel
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA; Graduate Program in Neurosciences, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA
| | - Nevin Varghese
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Allen B Reitz
- Fox Chase Chemical Diversity Center, Inc., Doylestown, PA 18902, USA
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104-6391, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854-8082, USA.
| |
Collapse
|
35
|
Choi JE, Lee JJ, Kang W, Kim HJ, Cho JH, Han PL, Lee KJ. Proteomic Analysis of Hippocampus in a Mouse Model of Depression Reveals Neuroprotective Function of Ubiquitin C-terminal Hydrolase L1 (UCH-L1) via Stress-induced Cysteine Oxidative Modifications. Mol Cell Proteomics 2018; 17:1803-1823. [PMID: 29959188 PMCID: PMC6126396 DOI: 10.1074/mcp.ra118.000835] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 06/20/2018] [Indexed: 01/08/2023] Open
Abstract
Chronic physical restraint stress increases oxidative stress in the brain, and dysregulation of oxidative stress can be one of the causes of major depressive disorder. To understand the underlying mechanisms, we undertook a systematic proteomic analysis of hippocampus in a chronic restraint stress mouse model of depression. Combining two-dimensional gel electrophoresis (2D-PAGE) for protein separation with nanoUPLC-ESI-q-TOF tandem mass spectrometry, we identified sixty-three protein spots that changed in the hippocampus of mice subjected to chronic restraint stress. We identified and classified the proteins that changed after chronic stress, into three groups respectively functioning in neural plasticity, metabolic processes and protein aggregation. Of these, 5 proteins including ubiquitin C-terminal hydrolase L1 (UCH-L1), dihydropyrimidinase-related protein 2 (DPYL2), haloacid dehalogenase-like hydrolase domain-containing protein 2 (HDHD2), actin-related protein 2/3 complex subunit 5 (ARPC5) and peroxiredoxin-2 (PRDX2), showed pI shifts attributable to post-translational modifications. Further analysis indicated that UCH-L1 underwent differential oxidations of 2 cysteine residues following chronic stress. We investigated whether the oxidized form of UCH-L1 plays a role in stressed hippocampus, by comparing the effects of UCH-L1 and its Cys mutants on hippocampal cell line HT-22 in response to oxidative stress. This study demonstrated that UCH-L1 wild-type and cysteine to aspartic acid mutants, but not its cysteine to serine mutants, afforded neuroprotective effects against oxidative stress; there were no discernible differences between wild-type UCH-L1 and its mutants in the absence of oxidative stress. These findings suggest that cysteine oxidative modifications of UCH-L1 in the hippocampus play key roles in neuroprotection against oxidative stress caused in major depressive disorder.
Collapse
Affiliation(s)
- Jung-Eun Choi
- From the ‡College of Pharmacy and Graduate School of Pharmaceutical Sciences, and
| | - Jae-Jin Lee
- From the ‡College of Pharmacy and Graduate School of Pharmaceutical Sciences, and
| | - Wonmo Kang
- From the ‡College of Pharmacy and Graduate School of Pharmaceutical Sciences, and
| | - Hyun Jung Kim
- From the ‡College of Pharmacy and Graduate School of Pharmaceutical Sciences, and
| | - Jin-Hwan Cho
- From the ‡College of Pharmacy and Graduate School of Pharmaceutical Sciences, and
| | - Pyung-Lim Han
- §Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, Korea 03760
| | - Kong-Joo Lee
- From the ‡College of Pharmacy and Graduate School of Pharmaceutical Sciences, and
| |
Collapse
|
36
|
Svane KC, Asis EK, Omelchenko A, Kunnath AJ, Brzustowicz LM, Silverstein SM, Firestein BL. d-Serine administration affects nitric oxide synthase 1 adaptor protein and DISC1 expression in sex-specific manner. Mol Cell Neurosci 2018; 89:20-32. [PMID: 29601869 DOI: 10.1016/j.mcn.2018.03.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/23/2018] [Accepted: 03/25/2018] [Indexed: 01/19/2023] Open
Abstract
Antipsychotic medications are inefficient at treating symptoms of schizophrenia (SCZ), and N-methyl d-aspartate receptor (NMDAR) agonists are potential therapeutic alternatives. As such, these agonists may act on different pathways and proteins altered in the brains of patients with SCZ than do antipsychotic medications. Here, we investigate the effects of administration of the antipsychotic haloperidol and NMDAR agonist d-serine on function and expression of three proteins that play significant roles in SCZ: nitric oxide synthase 1 adaptor protein (NOS1AP), dopamine D2 (D2) receptor, and disrupted in schizophrenia 1 (DISC1). We administered haloperidol or d-serine to male and female Sprague Dawley rats via intraperitoneal injection for 12 days and subsequently examined cortical expression of NOS1AP, D2 receptor, and DISC1. We found sex-specific effects of haloperidol and d-serine treatment on the expression of these proteins. Haloperidol significantly reduced expression of D2 receptor in male, but not female, rats. Conversely, d-serine reduced expression of NOS1AP in male rats and did not affect D2 receptor expression. d-serine treatment also reduced expression of DISC1 in male rats and increased DISC1 expression in female rats. As NOS1AP is overexpressed in the cortex of patients with SCZ and negatively regulates NMDAR signaling, we subsequently examined whether treatment with antipsychotics or NMDAR agonists can reverse the detrimental effects of NOS1AP overexpression in vitro as previously reported by our group. NOS1AP overexpression promotes reduced dendrite branching in vitro, and as such, we treated cortical neurons overexpressing NOS1AP with different antipsychotics (haloperidol, clozapine, fluphenazine) or d-serine for 24 h and determined the effects of these drugs on NOS1AP expression and dendrite branching. While antipsychotics did not affect NOS1AP protein expression or dendrite branching in vitro, d-serine reduced NOS1AP expression and rescued NOS1AP-mediated reductions in dendrite branching. Taken together, our data suggest that d-serine influences the function and expression of NOS1AP, D2 receptor, and DISC1 in a sex-specific manner and reverses the effects of NOS1AP overexpression on dendrite morphology.
Collapse
Affiliation(s)
- Kirsten C Svane
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA; Neuroscience Graduate Program, Rutgers, The State University of New Jersey, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Ericka-Kate Asis
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Anton Omelchenko
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA; Neuroscience Graduate Program, Rutgers, The State University of New Jersey, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Ansley J Kunnath
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Linda M Brzustowicz
- Department of Genetics, Rutgers, The State University of New Jersey, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Steven M Silverstein
- Division of Schizophrenia Research, Rutgers University Behavioral Health Care, 671 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
37
|
Zuccarini M, Giuliani P, Frinchi M, Mudò G, Serio RM, Belluardo N, Buccella S, Carluccio M, Condorelli DF, Caciagli F, Ciccarelli R, Di Iorio P. Uncovering the Signaling Pathway behind Extracellular Guanine-Induced Activation of NO System: New Perspectives in Memory-Related Disorders. Front Pharmacol 2018; 9:110. [PMID: 29515443 PMCID: PMC5826394 DOI: 10.3389/fphar.2018.00110] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/31/2018] [Indexed: 12/31/2022] Open
Abstract
Mounting evidence suggests that the guanine-based purines stand out as key player in cell metabolism and in several models of neurodegenerative disorders, such as Parkinson's and Alzheimer's diseases. Guanosine (GUO) and guanine (GUA) are extracellular signaling molecules derived from the breakdown of the correspondent nucleotide, GTP, and their intracellular and extracellular levels are regulated by the fine-tuned activity of two major enzymes, purine nucleoside phosphorylase (PNP) and guanine deaminase (GDA). Noteworthy, GUO and GUA, seem to play opposite roles in the modulation of cognitive functions, such as learning and memory. Indeed GUO, despite exerting neuroprotective, anti-apoptotic and neurotrophic effects, causes a decay of cognitive activities, whereas GUA administration in rats results in working memory improvement (prevented by L-NAME pre-treatment). This study was designed to investigate, in a model of SH-SY5Y neuroblastoma cell line, the signal transduction pathway activated by extracellular GUA. Altogether, our results showed that: (i) in addition to an enhanced phosphorylation of ASK1, p38 and JNK, likely linked to a non-massive and transient ROS production, the PKB/NO/sGC/cGMP/PKG/ERK cascade seems to be the main signaling pathway elicited by extracellular GUA; (ii) the activation of this pathway occurs in a pertussis-toxin sensitive manner, thus suggesting the involvement of a putative G protein coupled receptor; (iii) the GUA-induced NO production, strongly reduced by cell pre-treatment with L-NAME, is negatively modulated by the EPAC-cAMP-CaMKII pathway, which causes the over-expression of GDA that, in turn, reduces the levels of GUA. These molecular mechanisms activated by GUA may be useful to support our previous observation showing that GUA improves learning and memory functions through the stimulation of NO signaling pathway, and underscore the therapeutic potential of oral administration of guanine for treating memory-related disorders.
Collapse
Affiliation(s)
- Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Patricia Giuliani
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Monica Frinchi
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Giuseppa Mudò
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Rosa Maria Serio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Natale Belluardo
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Silvana Buccella
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Marzia Carluccio
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | | | - Francesco Caciagli
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Renata Ciccarelli
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| |
Collapse
|
38
|
A Novel Short Isoform of Cytosolic PSD-95 Interactor (Cypin) Regulates Neuronal Development. Mol Neurobiol 2018; 55:6269-6281. [PMID: 29294243 DOI: 10.1007/s12035-017-0849-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/19/2017] [Indexed: 10/18/2022]
Abstract
The guanine deaminase cypin (cytosolic PSD-95 interactor) binds to PSD-95 (postsynaptic density protein 95) and regulates dendrite branching by promoting microtubule polymerization. Here, we identify a novel short isoform of cypin, termed cypinS, which is expressed in mouse and human, but not rat, tissues. Cypin and cypinS mRNA and protein levels peak at P7 and P14 in the mouse brain, suggesting a role for these isoforms during development. Interestingly, although cypinS lacks guanine deaminase activity, overexpression of cypinS increases dendrite branching. This increase occurs further away from soma than do increases resulting from overexpression of cypin. In contrast, overexpression of cypin, but not cypinS, decreases dendritic spine density and maturity. This suggests that changes to spines, but not to dendrites, may be dependent on guanine deaminase activity. Furthermore, overexpression of either cypin or cypinS increases miniature excitatory postsynaptic current (mEPSC) frequency, pointing to a presynaptic role for both isoforms. Interestingly, overexpression of cypinS results in a significantly greater increase in frequency than does overexpression of cypin. Thus, cypin and cypinS play distinct roles in neuronal development.
Collapse
|
39
|
Gaded V, Anand R. Nucleobase deaminases: a potential enzyme system for new therapies. RSC Adv 2018; 8:23567-23577. [PMID: 35540270 PMCID: PMC9081823 DOI: 10.1039/c8ra04112a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 06/11/2018] [Indexed: 11/21/2022] Open
Abstract
This review presents an overview of the structure, function and mechanism of CDA deaminases and their potential as enzyme systems for development of new antimicrobial therapies.
Collapse
Affiliation(s)
- Vandana Gaded
- Department of Chemistry
- Indian Institute of Technology Bombay
- Mumbai
- India
| | - Ruchi Anand
- Department of Chemistry
- Indian Institute of Technology Bombay
- Mumbai
- India
| |
Collapse
|
40
|
Distinct effects on the dendritic arbor occur by microbead versus bath administration of brain-derived neurotrophic factor. Cell Mol Life Sci 2017; 74:4369-4385. [PMID: 28698933 DOI: 10.1007/s00018-017-2589-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 06/15/2017] [Accepted: 07/06/2017] [Indexed: 12/18/2022]
Abstract
Proper communication among neurons depends on an appropriately formed dendritic arbor, and thus, aberrant changes to the arbor are implicated in many pathologies, ranging from cognitive disorders to neurodegenerative diseases. Due to the importance of dendritic shape to neuronal network function, the morphology of dendrites is tightly controlled and is influenced by both intrinsic and extrinsic factors. In this work, we examine how brain-derived neurotrophic factor (BDNF), one of the most well-studied extrinsic regulators of dendritic branching, affects the arbor when it is applied locally via microbeads to cultures of hippocampal neurons. We found that local application of BDNF increases both proximal and distal branching in a time-dependent manner and that local BDNF application attenuates pruning of dendrites that occurs with neuronal maturation. Additionally, we examined whether cytosolic PSD-95 interactor (cypin), an intrinsic regulator of dendritic branching, plays a role in these changes and found strong evidence for the involvement of cypin in BDNF-promoted increases in dendrites after 24 but not 48 h of application. This current study extends our previous work in which we found that bath application of BDNF for 72 h, but not shorter times, increases proximal dendrite branching and that this increase occurs through transcriptional regulation of cypin. Moreover, this current work illustrates how dendritic branching is regulated differently by the same growth factor depending on its spatial localization, suggesting a novel pathway for modulation of dendritic branching locally.
Collapse
|
41
|
Edmiston E, Ashwood P, Van de Water J. Autoimmunity, Autoantibodies, and Autism Spectrum Disorder. Biol Psychiatry 2017; 81:383-390. [PMID: 28340985 PMCID: PMC5373490 DOI: 10.1016/j.biopsych.2016.08.031] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/27/2016] [Accepted: 08/24/2016] [Indexed: 12/15/2022]
Abstract
Auism spectrum disorder (ASD) now affects one in 68 births in the United States and is the fastest growing neurodevelopmental disability worldwide. Alarmingly, for the majority of cases, the causes of ASD are largely unknown, but it is becoming increasingly accepted that ASD is no longer defined simply as a behavioral disorder, but rather as a highly complex and heterogeneous biological disorder. Although research has focused on the identification of genetic abnormalities, emerging studies increasingly suggest that immune dysfunction is a viable risk factor contributing to the neurodevelopmental deficits observed in ASD. This review summarizes the investigations implicating autoimmunity and autoantibodies in ASD.
Collapse
Affiliation(s)
- Elizabeth Edmiston
- Division of Rheumatology/Allergy and Clinical Immunology, University of California, Davis, Davis, California; The M.I.N.D. Institute, University of California, Davis, Davis, California
| | - Paul Ashwood
- The M.I.N.D. Institute, University of California, Davis, Davis, California; NIEHS Center for Children's Environmental Health, University of California, Davis, Davis, California; Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California
| | - Judy Van de Water
- Division of Rheumatology/Allergy and Clinical Immunology, University of California, Davis, Davis, California; The M.I.N.D. Institute, University of California, Davis, Davis, California; NIEHS Center for Children's Environmental Health, University of California, Davis, Davis, California.
| |
Collapse
|
42
|
Bastian TW, von Hohenberg WC, Mickelson DJ, Lanier LM, Georgieff MK. Iron Deficiency Impairs Developing Hippocampal Neuron Gene Expression, Energy Metabolism, and Dendrite Complexity. Dev Neurosci 2016; 38:264-276. [PMID: 27669335 DOI: 10.1159/000448514] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 07/05/2016] [Indexed: 12/28/2022] Open
Abstract
Iron deficiency (ID), with and without anemia, affects an estimated 2 billion people worldwide. ID is particularly deleterious during early-life brain development, leading to long-term neurological impairments including deficits in hippocampus-mediated learning and memory. Neonatal rats with fetal/neonatal ID anemia (IDA) have shorter hippocampal CA1 apical dendrites with disorganized branching. ID-induced dendritic structural abnormalities persist into adulthood despite normalization of the iron status. However, the specific developmental effects of neuronal iron loss on hippocampal neuron dendrite growth and branching are unknown. Embryonic hippocampal neuron cultures were chronically treated with deferoxamine (DFO, an iron chelator) beginning at 3 days in vitro (DIV). Levels of mRNA for Tfr1 and Slc11a2, iron-responsive genes involved in iron uptake, were significantly elevated in DFO-treated cultures at 11DIV and 18DIV, indicating a degree of neuronal ID similar to that seen in rodent ID models. DFO treatment decreased mRNA levels for genes indexing dendritic and synaptic development (i.e. BdnfVI,Camk2a,Vamp1,Psd95,Cfl1, Pfn1,Pfn2, and Gda) and mitochondrial function (i.e. Ucp2,Pink1, and Cox6a1). At 18DIV, DFO reduced key aspects of energy metabolism including basal respiration, maximal respiration, spare respiratory capacity, ATP production, and glycolytic rate, capacity, and reserve. Sholl analysis revealed a significant decrease in distal dendritic complexity in DFO-treated neurons at both 11DIV and 18DIV. At 11DIV, the length of primary dendrites and the number and length of branches in DFO-treated neurons were reduced. By 18DIV, partial recovery of the dendritic branch number in DFO-treated neurons was counteracted by a significant reduction in the number and length of primary dendrites and the length of branches. Our findings suggest that early neuronal iron loss, at least partially driven through altered mitochondrial function and neuronal energy metabolism, is responsible for the effects of fetal/neonatal ID and IDA on hippocampal neuron dendritic and synaptic maturation. Impairments in these neurodevelopmental processes likely underlie the negative impact of early life ID and IDA on hippocampus-mediated learning and memory.
Collapse
Affiliation(s)
- Thomas W Bastian
- Department of Pediatrics and Center for Neurobehavioral Development, School of Medicine, Minneapolis, Minn., USA
| | | | | | | | | |
Collapse
|
43
|
Gonzalez A, Moya-Alvarado G, Gonzalez-Billaut C, Bronfman FC. Cellular and molecular mechanisms regulating neuronal growth by brain-derived neurotrophic factor. Cytoskeleton (Hoboken) 2016; 73:612-628. [PMID: 27223597 DOI: 10.1002/cm.21312] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/20/2016] [Accepted: 05/23/2016] [Indexed: 12/31/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) and its receptors TrkB and p75 regulate dendritic and axonal growth during development and maintenance of the mature nervous system; however, the cellular and molecular mechanisms underlying this process are not fully understood. In recent years, several advances have shed new light on the processes behind the regulation of BDNF-mediated structural plasticity including control of neuronal transcription, local translation of proteins, and regulation of cytoskeleton and membrane dynamics. In this review, we summarize recent advances in the field of BDNF signaling in neurons to induce neuronal growth. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Andres Gonzalez
- MINREB and Center for Ageing and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Guillermo Moya-Alvarado
- MINREB and Center for Ageing and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christian Gonzalez-Billaut
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile and Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Francisca C Bronfman
- MINREB and Center for Ageing and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
44
|
Jayaraman A, Thandeeswaran M, Priyadarsini U, Sabarathinam S, Nawaz KAA, Palaniswamy M. Characterization of unexplored amidohydrolase enzyme-pterin deaminase. Appl Microbiol Biotechnol 2016; 100:4779-89. [PMID: 27094187 DOI: 10.1007/s00253-016-7513-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/28/2016] [Accepted: 03/30/2016] [Indexed: 11/30/2022]
Abstract
Pterin deaminase is an amidohydrolase enzyme hydrolyzing pteridines to form lumazine derivatives and ammonia. The enzyme captured the attention of scientists as early as 1959 and had been patented for its application as an anticancer agent. It is ubiquitously present in prokaryotes and has been reported in some eukaryotes such as honey bee, silkworm and rats. The enzyme has been observed to have a spectrum of substrates with the formation of respective lumazines. The role of the substrates of the enzyme in various metabolic pathways warrants a significant role in the biological activity of both prokaryotes and eukaryotes. Even though the functions of the enzyme have been explored in prokaryotes, their niche in the eukaryotic system is not clear. There is very few information on the structural and functional properties of the enzyme. This review has been congregated to emphasize the significance of pterin deaminase and analyzes the lacunae in understanding the biological characters of the enzyme.
Collapse
Affiliation(s)
- Angayarkanni Jayaraman
- Cancer Therapeutics Lab, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, Tamilnadu, India.
| | - Murugesan Thandeeswaran
- Cancer Therapeutics Lab, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, Tamilnadu, India
| | | | - Shanmugam Sabarathinam
- Cancer Therapeutics Lab, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, Tamilnadu, India
| | - K A Ayub Nawaz
- Cancer Therapeutics Lab, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, Tamilnadu, India
| | - Muthusamy Palaniswamy
- Department of Microbiology, Karpagam University, Coimbatore, 641021, Tamilnadu, India
| |
Collapse
|
45
|
Posttranslational Modifications Regulate the Postsynaptic Localization of PSD-95. Mol Neurobiol 2016; 54:1759-1776. [PMID: 26884267 DOI: 10.1007/s12035-016-9745-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/22/2016] [Indexed: 01/08/2023]
Abstract
The postsynaptic density (PSD) consists of a lattice-like array of interacting proteins that organizes and stabilizes synaptic receptors, ion channels, structural proteins, and signaling molecules required for normal synaptic transmission and synaptic function. The scaffolding and hub protein postsynaptic density protein-95 (PSD-95) is a major element of central chemical synapses and interacts with glutamate receptors, cell adhesion molecules, and cytoskeletal elements. In fact, PSD-95 can regulate basal synaptic stability as well as the activity-dependent structural plasticity of the PSD and, therefore, of the excitatory chemical synapse. Several studies have shown that PSD-95 is highly enriched at excitatory synapses and have identified multiple protein structural domains and protein-protein interactions that mediate PSD-95 function and trafficking to the postsynaptic region. PSD-95 is also a target of several signaling pathways that induce posttranslational modifications, including palmitoylation, phosphorylation, ubiquitination, nitrosylation, and neddylation; these modifications determine the synaptic stability and function of PSD-95 and thus regulate the fates of individual dendritic spines in the nervous system. In the present work, we review the posttranslational modifications that regulate the synaptic localization of PSD-95 and describe their functional consequences. We also explore the signaling pathways that induce such changes.
Collapse
|
46
|
Vacca M, Tripathi KP, Speranza L, Aiese Cigliano R, Scalabrì F, Marracino F, Madonna M, Sanseverino W, Perrone-Capano C, Guarracino MR, D'Esposito M. Effects of Mecp2 loss of function in embryonic cortical neurons: a bioinformatics strategy to sort out non-neuronal cells variability from transcriptome profiling. BMC Bioinformatics 2016; 17 Suppl 2:14. [PMID: 26821710 PMCID: PMC4959389 DOI: 10.1186/s12859-015-0859-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Mecp2 null mice model Rett syndrome (RTT) a human neurological disorder affecting females after apparent normal pre- and peri-natal developmental periods. Neuroanatomical studies in cerebral cortex of RTT mouse models revealed delayed maturation of neuronal morphology and autonomous as well as non-cell autonomous reduction in dendritic complexity of postnatal cortical neurons. However, both morphometric parameters and high-resolution expression profile of cortical neurons at embryonic developmental stage have not yet been studied. Here we address these topics by using embryonic neuronal primary cultures from Mecp2 loss of function mouse model. Results We show that embryonic primary cortical neurons of Mecp2 null mice display reduced neurite complexity possibly reflecting transcriptional changes. We used RNA-sequencing coupled with a bioinformatics comparative approach to identify and remove the contribution of variable and hard to quantify non-neuronal brain cells present in our in vitro cell cultures. Conclusions Our results support the need to investigate both Mecp2 morphological as well as molecular effect in neurons since prenatal developmental stage, long time before onset of Rett symptoms. Electronic supplementary material The online version of this article (doi:10.1186/s12859-015-0859-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marcella Vacca
- Institute of Genetics and Biophysics "A. Buzzati Traverso", National Research Council (CNR)-80131, Naples, Italy.
| | - Kumar Parijat Tripathi
- Laboratory for Genomics, Transcriptomics and Proteomics (LAB-GTP), High Performance Computing and Networking Institute (ICAR), National Research Council (CNR)-80131, Naples, Italy.
| | - Luisa Speranza
- Institute of Genetics and Biophysics "A. Buzzati Traverso", National Research Council (CNR)-80131, Naples, Italy.
| | | | | | | | | | - Walter Sanseverino
- Sequentia Biotech SL, Calle Comte D'Urgell, 240 08036, Barcelona, Spain.
| | - Carla Perrone-Capano
- Institute of Genetics and Biophysics "A. Buzzati Traverso", National Research Council (CNR)-80131, Naples, Italy. .,Department of Pharmacy, University of Naples Federico II, Naples, Italy.
| | - Mario Rosario Guarracino
- Laboratory for Genomics, Transcriptomics and Proteomics (LAB-GTP), High Performance Computing and Networking Institute (ICAR), National Research Council (CNR)-80131, Naples, Italy.
| | - Maurizio D'Esposito
- Institute of Genetics and Biophysics "A. Buzzati Traverso", National Research Council (CNR)-80131, Naples, Italy. .,IRCCS Neuromed, via dell'Elettronica, Pozzilli (Is), Italy.
| |
Collapse
|
47
|
O'Neill KM, Akum BF, Dhawan ST, Kwon M, Langhammer CG, Firestein BL. Assessing effects on dendritic arborization using novel Sholl analyses. Front Cell Neurosci 2015; 9:285. [PMID: 26283921 PMCID: PMC4519774 DOI: 10.3389/fncel.2015.00285] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/13/2015] [Indexed: 01/12/2023] Open
Abstract
Determining the shape of cell-specific dendritic arbors is a tightly regulated process that occurs during development. When this regulation is aberrant, which occurs during disease or injury, alterations in dendritic shape result in changes to neural circuitry. There has been significant progress on characterizing extracellular and intrinsic factors that regulate dendrite number by our laboratory and others. Generally, changes to the dendritic arbor are assessed by Sholl analysis or simple dendrite counting. However, we have found that this general method often overlooks local changes to the arbor. Previously, we developed a program (titled Bonfire) to facilitate digitization of neurite morphology and subsequent Sholl analysis and to assess changes to root, intermediate, and terminal neurites. Here, we apply these different Sholl analyses, and a novel Sholl analysis, to uncover previously unknown changes to the dendritic arbor when we overexpress an important regulator of dendrite branching, cytosolic PSD-95 interactor (cypin), at two developmental time points. Our results suggest that standard Sholl analysis and simple dendrite counting are not sufficient for uncovering local changes to the dendritic arbor.
Collapse
Affiliation(s)
- Kate M O'Neill
- Department of Cell Biology and Neuroscience, Rutgers University Piscataway, NJ, USA ; Graduate Program in Biomedical Engineering, Rutgers University Piscataway, NJ, USA
| | - Barbara F Akum
- Department of Cell Biology and Neuroscience, Rutgers University Piscataway, NJ, USA
| | - Survandita T Dhawan
- Department of Cell Biology and Neuroscience, Rutgers University Piscataway, NJ, USA
| | - Munjin Kwon
- Department of Cell Biology and Neuroscience, Rutgers University Piscataway, NJ, USA
| | | | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers University Piscataway, NJ, USA
| |
Collapse
|
48
|
Wierzchowski J, Antosiewicz JM, Shugar D. 8-Azapurines as isosteric purine fluorescent probes for nucleic acid and enzymatic research. MOLECULAR BIOSYSTEMS 2015; 10:2756-74. [PMID: 25124808 DOI: 10.1039/c4mb00233d] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The 8-azapurines, and their 7-deaza and 9-deaza congeners, represent a unique class of isosteric (isomorphic) analogues of the natural purines, frequently capable of substituting for the latter in many biochemical processes. Particularly interesting is their propensity to exhibit pH-dependent room-temperature fluorescence in aqueous medium, and in non-polar media. We herein review the physico-chemical properties of this class of compounds, with particular emphasis on the fluorescence emission properties of their neutral and/or ionic species, which has led to their widespread use as fluorescent probes in enzymology, including enzymes involved in purine metabolism, agonists/antagonists of adenosine receptors, mechanisms of catalytic RNAs, RNA editing, etc. They are also exceptionally useful fluorescent probes for analytical and clinical applications in crude cell homogenates.
Collapse
Affiliation(s)
- Jacek Wierzchowski
- Department of Biophysics, University of Varmia & Masuria, Oczapowskiego 4, 10-719 Olsztyn, Poland.
| | | | | |
Collapse
|
49
|
Zhang YW, Zheng Y, Wang JZ, LU XX, Wang Z, Chen LB, Guan XX, Tong JD. Integrated analysis of DNA methylation and mRNA expression profiling reveals candidate genes associated with cisplatin resistance in non-small cell lung cancer. Epigenetics 2014; 9:896-909. [PMID: 24699858 PMCID: PMC4065187 DOI: 10.4161/epi.28601] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/15/2014] [Accepted: 03/19/2014] [Indexed: 01/07/2023] Open
Abstract
DNA methylation plays a critical role during the development of acquired chemoresistance. The aim of this study was to identify candidate DNA methylation drivers of cisplatin (DDP) resistance in non-small cell lung cancer (NSCLC). The A549/DDP cell line was established by continuous exposure of A549 cells to increasing concentrations of DDP. Gene expression and methylation profiling were determined by high-throughput microarrays. Relationship of methylation status and DDP response was validated in primary tumor cell culture and the Cancer Genome Atlas (TCGA) samples. Cell proliferation, apoptosis, cell cycle, and response to DDP were determined in vitro and in vivo. A total of 372 genes showed hypermethylation and downregulation in A549/DDP cells, and these genes were involved in most fundamental biological processes. Ten candidate genes (S100P, GDA, WISP2, LOXL1, TIMP4, ICAM1, CLMP, HSP8, GAS1, BMP2) were selected, and exhibited varying degrees of association with DDP resistance. Low dose combination of 5-aza-2'-deoxycytidine (5-Aza-dC) and trichostatin A (TSA) reversed drug resistance of A549/DDP cells in vitro and in vivo, along with demethylation and restoration of expression of candidate genes (GAS1, TIMP4, ICAM1 and WISP2). Forced expression of GAS1 in A549/DDP cells by gene transfection contributed to increased sensitivity to DDP, proliferation inhibition, cell cycle arrest, apoptosis enhancement, and in vivo growth retardation. Together, our study demonstrated that a panel of candidate genes downregulated by DNA methylation induced DDP resistance in NSCLC, and showed that epigenetic therapy resensitized cells to DDP.
Collapse
Affiliation(s)
- You-Wei Zhang
- Department of Oncology; Jinling Hospital; Medical School of Nanjing University; Nanjing, PR China
- Department of Oncology; Yangzhou No. 1 Hospital; The Second Clinical School of Yangzhou University; Yangzhou, PR China
- Department of Oncology; Affiliated Xuzhou Central Hospital; Xuzhou Medical College; Xuzhou, PR China
| | - Yun Zheng
- Department of Oncology; Jinling Hospital; Medical School of Nanjing University; Nanjing, PR China
| | - Jing-Zi Wang
- Department of Oncology; Jinling Hospital; Medical School of Nanjing University; Nanjing, PR China
| | - Xiao-Xia LU
- Department of Oncology; Yangzhou No. 1 Hospital; The Second Clinical School of Yangzhou University; Yangzhou, PR China
| | - Zhu Wang
- Department of Oncology; Yangzhou No. 1 Hospital; The Second Clinical School of Yangzhou University; Yangzhou, PR China
| | - Long-Bang Chen
- Department of Oncology; Jinling Hospital; Medical School of Nanjing University; Nanjing, PR China
| | - Xiao-Xiang Guan
- Department of Oncology; Jinling Hospital; Medical School of Nanjing University; Nanjing, PR China
| | - Jian-Dong Tong
- Department of Oncology; Yangzhou No. 1 Hospital; The Second Clinical School of Yangzhou University; Yangzhou, PR China
| |
Collapse
|
50
|
Fuller HR, Hurtado ML, Wishart TM, Gates MA. The rat striatum responds to nigro-striatal degeneration via the increased expression of proteins associated with growth and regeneration of neuronal circuitry. Proteome Sci 2014; 12:20. [PMID: 24834013 PMCID: PMC4021461 DOI: 10.1186/1477-5956-12-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/17/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Idiopathic Parkinson's disease is marked by degeneration of dopamine neurons projecting from the substantia nigra to the striatum. Although proteins expressed by the target striatum can positively affect the viability and growth of dopaminergic neurons, very little is known about the molecular response of the striatum as nigro-striatal denervation progresses. Here, iTRAQ labelling and MALDI TOF/TOF mass spectrometry have been used to quantitatively compare the striatal proteome of rats before, during, and after 6-OHDA induced dopamine denervation. RESULTS iTRAQ analysis revealed the differential expression of 50 proteins at 3 days, 26 proteins at 7 days, and 34 proteins at 14 days post-lesioning, compared to the unlesioned striatum. While the denervated striatum showed a reduced expression of proteins associated with the loss of dopaminergic input (e.g., TH and DARPP-32), there was an increased expression of proteins associated with regeneration and growth of neurites (e.g., GFAP). In particular, the expression of guanine deaminase (GDA, cypin) - a protein known to be involved in dendritic branching - was significantly increased in the striatum at 3, 7 and 14 days post-lesioning (a finding verified by immunohistochemistry). CONCLUSIONS Together, these findings provide evidence to suggest that the response of the normal mammalian striatum to nigro-striatal denervation includes the increased expression of proteins that may have the capacity to facilitate repair and growth of neuronal circuitry.
Collapse
Affiliation(s)
- Heidi R Fuller
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK,Keele University, Institute for Science and Technology in Medicine, Department of Life Sciences, Huxley Building, Keele ST5 5BG, UK
| | - Maica Llavero Hurtado
- Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Thomas M Wishart
- Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK,Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| | - Monte A Gates
- Keele University, Institute for Science and Technology in Medicine, Department of Life Sciences, Huxley Building, Keele ST5 5BG, UK
| |
Collapse
|