1
|
Aderibigbe O, Wood LB, Margulies SS. Cyclosporine A Accelerates Neurorecovery Transcriptional Trajectory in a Swine Model of Diffuse Traumatic Brain Injury. Int J Mol Sci 2025; 26:3531. [PMID: 40331981 PMCID: PMC12026708 DOI: 10.3390/ijms26083531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/04/2025] [Accepted: 04/05/2025] [Indexed: 05/08/2025] Open
Abstract
Mild traumatic brain injury (mTBI) is a leading cause of morbidity in children with both short- and long-term neurological, cognitive, cerebrovascular, and emotional deficits. These deficits have been attributed to ongoing pathophysiological cascades that occur acutely and persist post-injury. Given our limited understanding of the transcriptional changes associated with these pathophysiological cascades, we studied formalin-fixed paraffin-embedded (FFPE) tissues from the frontal cortex (FC) and the hippocampus + amygdala (H&A) regions of swine (N = 40) after a sagittal rapid non-impact head rotation (RNR). We then sequenced RNA to define transcriptional changes at 1 day and 1 week after injury and investigated the protective influence of cyclosporine A (CsA) treatment. Differentially expressed genes (DEGs) were classified into five temporal patterns (Early, Transient, Persistent, Intensified, Delayed, or Late). DEGs were more abundant at 1 week than 1 day. Shared significant gene ontology annotations in both regions included terms associated with neuronal distress at 1 day and neurorecovery at 1 week. CsA (20 mg/kg/day) infused for 1 day (beginning at 6 h after injury) accelerated 466 DEGs in the FC and 2794 DEGs in the H&A, such that the CsA-treated transcriptional profile was associated with neurorecovery. Overall, our data reveal the effects of anatomic region and elapsed time on gene expression post-mTBI and motivate future studies of CsA treatment.
Collapse
Affiliation(s)
- Oluwagbemisola Aderibigbe
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA;
| | - Levi B. Wood
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA;
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Susan S. Margulies
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA;
| |
Collapse
|
2
|
Miyata S, Tsuda M, Mitsui S. Overexpression of Motopsin, an Extracellular Serine Protease Related to Intellectual Disability, Promotes Adult Neurogenesis and Neuronal Responsiveness in the Dentate Gyrus. Mol Neurobiol 2024; 61:4929-4948. [PMID: 38153682 DOI: 10.1007/s12035-023-03890-y] [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: 05/24/2023] [Accepted: 12/18/2023] [Indexed: 12/29/2023]
Abstract
Motopsin, a serine protease encoded by PRSS12, is secreted by neuronal cells into the synaptic clefts in an activity-dependent manner, where it induces synaptogenesis by modulating Na+/K+-ATPase activity. In humans, motopsin deficiency leads to severe intellectual disability and, in mice, it disturbs spatial memory and social behavior. In this study, we investigated mice that overexpressed motopsin in the forebrain using the Tet-Off system (DTG-OE mice). The elevated agrin cleavage or the reduced Na+/K+-ATPase activity was not detected. However, motopsin overexpression led to a reduction in spine density in hippocampal CA1 basal dendrites. While motopsin overexpression decreased the ratio of mature mushroom spines in the DG, it increased the ratio of immature thin spines in CA1 apical dendrites. Female DTG-OE mice showed elevated locomotor activity in their home cages. DTG-OE mice showed aberrant behaviors, such as delayed latency to the target hole in the Barnes maze test and prolonged duration of sniffing objects in the novel object recognition test (NOR), although they retained memory comparable to that of TRE-motopsin littermates, which normally express motopsin. After NOR, c-Fos-positive cells increased in the dentate gyrus (DG) of DTG-OE mice compared with that of DTG-SO littermates, in which motopsin overexpression was suppressed by the administration of doxycycline, and TRE-motopsin littermates. Notably, the numbers of doublecortin- and 5-bromo-2'-deoxyuridine-labeled cells significantly increased in the DG of DTG-OE mice, suggesting increased adult neurogenesis. Importantly, our results revealed a new function in addition to modulating neuronal responsiveness and spine morphology in the DG: the regulation of neurogenesis.
Collapse
Affiliation(s)
- Shiori Miyata
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa, Maebashi, Gunma, 371-8514, Japan
| | - Masayuki Tsuda
- Division of Laboratory Animal Science, Science Research Center, Kochi Medical School, Kochi University, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Shinichi Mitsui
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa, Maebashi, Gunma, 371-8514, Japan.
| |
Collapse
|
3
|
Kathuria A, Lopez-Lengowski K, Watmuff B, Karmacharya R. Morphological and transcriptomic analyses of stem cell-derived cortical neurons reveal mechanisms underlying synaptic dysfunction in schizophrenia. Genome Med 2023; 15:58. [PMID: 37507766 PMCID: PMC10375745 DOI: 10.1186/s13073-023-01203-5] [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: 08/14/2022] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Postmortem studies in schizophrenia consistently show reduced dendritic spines in the cerebral cortex but the mechanistic underpinnings of these deficits remain unknown. Recent genome-wide association studies and exome sequencing investigations implicate synaptic genes and processes in the disease biology of schizophrenia. METHODS We generated human cortical pyramidal neurons by differentiating iPSCs of seven schizophrenia patients and seven healthy subjects, quantified dendritic spines and synapses in different cortical neuron subtypes, and carried out transcriptomic studies to identify differentially regulated genes and aberrant cellular processes in schizophrenia. RESULTS Cortical neurons expressing layer III marker CUX1, but not those expressing layer V marker CTIP2, showed significant reduction in dendritic spine density in schizophrenia, mirroring findings in postmortem studies. Transcriptomic experiments in iPSC-derived cortical neurons showed that differentially expressed genes in schizophrenia were enriched for genes implicated in schizophrenia in genome-wide association and exome sequencing studies. Moreover, most of the differentially expressed genes implicated in schizophrenia genetic studies had lower expression levels in schizophrenia cortical neurons. Network analysis of differentially expressed genes led to identification of NRXN3 as a hub gene, and follow-up experiments showed specific reduction of the NRXN3 204 isoform in schizophrenia neurons. Furthermore, overexpression of the NRXN3 204 isoform in schizophrenia neurons rescued the spine and synapse deficits in the cortical neurons while knockdown of NRXN3 204 in healthy neurons phenocopied spine and synapse deficits seen in schizophrenia cortical neurons. The antipsychotic clozapine increased expression of the NRXN3 204 isoform in schizophrenia cortical neurons and rescued the spine and synapse density deficits. CONCLUSIONS Taken together, our findings in iPSC-derived cortical neurons recapitulate cell type-specific findings in postmortem studies in schizophrenia and have led to the identification of a specific isoform of NRXN3 that modulates synaptic deficits in schizophrenia neurons.
Collapse
Affiliation(s)
- Annie Kathuria
- Harvard University, MGH Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN6, Boston, MA, 02114, USA
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Kara Lopez-Lengowski
- Harvard University, MGH Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN6, Boston, MA, 02114, USA
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Bradley Watmuff
- Harvard University, MGH Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN6, Boston, MA, 02114, USA
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Rakesh Karmacharya
- Harvard University, MGH Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN6, Boston, MA, 02114, USA.
- Chemical Biology Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Program in Neuroscience, Harvard University, Cambridge, MA, USA.
- Schizophrenia & Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA.
- Program in Chemical Biology, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
4
|
Wang YB, Song NN, Ding YQ, Zhang L. Neural plasticity and depression treatment. IBRO Neurosci Rep 2023; 14:160-184. [PMID: 37388497 PMCID: PMC10300479 DOI: 10.1016/j.ibneur.2022.09.001] [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: 05/31/2022] [Revised: 07/29/2022] [Accepted: 09/01/2022] [Indexed: 12/08/2022] Open
Abstract
Depression is one of the most common mental disorders, which can lead to a variety of emotional problems and even suicide at its worst. As this neuropsychiatric disorder causes the patients to suffer a lot and function poorly in everyday life, it is imposing a heavy burden on the affected families and the whole society. Several hypotheses have been proposed to elucidate the pathogenesis of depression, such as the genetic mutations, the monoamine hypothesis, the hypothalamic-pituitary-adrenal (HPA) axis hyperactivation, the inflammation and the neural plasticity changes. Among these models, neural plasticity can occur at multiple levels from brain regions, cells to synapses structurally and functionally during development and in adulthood. In this review, we summarize the recent progresses (especially in the last five years) on the neural plasticity changes in depression under different organizational levels and elaborate different treatments for depression by changing the neural plasticity. We hope that this review would shed light on the etiological studies for depression and on the development of novel treatments.
Collapse
Affiliation(s)
- Yu-Bing Wang
- Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center) and Department of Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai 200092, China
| | - Ning-Ning Song
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudfan University, Shanghai 200032, China
| | - Yu-Qiang Ding
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudfan University, Shanghai 200032, China
| | - Lei Zhang
- Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center) and Department of Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai 200092, China
| |
Collapse
|
5
|
Deconstruction of Neurotrypsin Reveals a Multi-factorially Regulated Activity Affecting Myotube Formation and Neuronal Excitability. Mol Neurobiol 2022; 59:7466-7485. [PMID: 36197591 PMCID: PMC9616769 DOI: 10.1007/s12035-022-03056-2] [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: 06/03/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022]
Abstract
Neurotrypsin (NT) is a highly specific nervous system multi-domain serine protease best known for its selective processing of the potent synaptic organizer agrin. Its enzymatic activity is thought to influence processes of synaptic plasticity, with its deregulation causing accelerated neuromuscular junction (NMJ) degeneration or contributing to forms of mental retardation. These biological effects are likely to stem from NT-based regulation of agrin signaling. However, dissecting the exact biological implications of NT-agrin interplay is difficult, due to the scarce molecular detail regarding NT activity and NT-agrin interactions. We developed a strategy to reliably produce and purify a catalytically competent engineered variant of NT called "NT-mini" and a library of C-terminal agrin fragments, with which we performed a thorough biochemical and biophysical characterization of NT enzyme functionality. We studied the regulatory effects of calcium ions and heparin, identified NT's heparin-binding domain, and discovered how zinc ions induce modulation of enzymatic activity. Additionally, we investigated myotube differentiation and hippocampal neuron excitability, evidencing a dose-dependent increase in neuronal activity alongside a negative impact on myoblast fusion when using the active NT enzyme. Collectively, our results provide in vitro and cellular foundations to unravel the molecular underpinnings and biological significance of NT-agrin interactions.
Collapse
|
6
|
Qiu WQ, Luo S, Ma SA, Saminathan P, Li H, Gunnersen JM, Gelbard HA, Hammond JW. The Sez6 Family Inhibits Complement by Facilitating Factor I Cleavage of C3b and Accelerating the Decay of C3 Convertases. Front Immunol 2021; 12:607641. [PMID: 33936031 PMCID: PMC8081827 DOI: 10.3389/fimmu.2021.607641] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 03/23/2021] [Indexed: 12/31/2022] Open
Abstract
The Sez6 family consists of Sez6, Sez6L, and Sez6L2. Its members are expressed throughout the brain and have been shown to influence synapse numbers and dendritic morphology. They are also linked to various neurological and psychiatric disorders. All Sez6 family members contain 2-3 CUB domains and 5 complement control protein (CCP) domains, suggesting that they may be involved in complement regulation. We show that Sez6 family members inhibit C3b/iC3b opsonization by the classical and alternative pathways with varying degrees of efficacy. For the classical pathway, Sez6 is a strong inhibitor, Sez6L2 is a moderate inhibitor, and Sez6L is a weak inhibitor. For the alternative pathway, the complement inhibitory activity of Sez6, Sez6L, and Sez6L2 all equaled or exceeded the activity of the known complement regulator MCP. Using Sez6L2 as the representative family member, we show that it specifically accelerates the dissociation of C3 convertases. Sez6L2 also functions as a cofactor for Factor I to facilitate the cleavage of C3b; however, Sez6L2 has no cofactor activity toward C4b. In summary, the Sez6 family are novel complement regulators that inhibit C3 convertases and promote C3b degradation.
Collapse
Affiliation(s)
- Wen Q Qiu
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Shaopeiwen Luo
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Stefanie A Ma
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Priyanka Saminathan
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Herman Li
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Jenny M Gunnersen
- Department of Anatomy and Neuroscience and The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Harris A Gelbard
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Jennetta W Hammond
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
7
|
Zhu L, Chen L, Xu P, Lu D, Dai S, Zhong L, Han Y, Zhang M, Xiao B, Chang L, Wu Q. Genetic and molecular basis of epilepsy-related cognitive dysfunction. Epilepsy Behav 2020; 104:106848. [PMID: 32028124 DOI: 10.1016/j.yebeh.2019.106848] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/06/2019] [Accepted: 12/06/2019] [Indexed: 02/02/2023]
Abstract
Epilepsy is a common neurological disease characterized by recurrent seizures. About 70 million people were affected by epilepsy or epileptic seizures. Epilepsy is a complicated complex or symptomatic syndromes induced by structural, functional, and genetic causes. Meanwhile, several comorbidities are accompanied by epileptic seizures. Cognitive dysfunction is a long-standing complication associated with epileptic seizures, which severely impairs quality of life. Although the definitive pathogenic mechanisms underlying epilepsy-related cognitive dysfunction remain unclear, accumulating evidence indicates that multiple risk factors are probably involved in the development and progression of cognitive dysfunction in patients with epilepsy. These factors include the underlying etiology, recurrent seizures or status epilepticus, structural damage that induced secondary epilepsy, genetic variants, and molecular alterations. In this review, we summarize several theories that may explain the genetic and molecular basis of epilepsy-related cognitive dysfunction.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China
| | - Lu Chen
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China
| | - Puying Xu
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China
| | - Di Lu
- Biomedicine Engineering Research Center, Kunming Medical University, 1168 Chun Rong West Road, Kunming, Yunnan 650500, PR China
| | - Shujuan Dai
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China
| | - Lianmei Zhong
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China
| | - Yanbing Han
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China
| | - Mengqi Zhang
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiang Ya Road, Changsha, Hunan 410008, PR China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiang Ya Road, Changsha, Hunan 410008, PR China
| | - Lvhua Chang
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China.
| | - Qian Wu
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China.
| |
Collapse
|
8
|
Guarino SR, Canciani A, Forneris F. Dissecting the Extracellular Complexity of Neuromuscular Junction Organizers. Front Mol Biosci 2020; 6:156. [PMID: 31998752 PMCID: PMC6966886 DOI: 10.3389/fmolb.2019.00156] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/13/2019] [Indexed: 12/20/2022] Open
Abstract
Synapse formation is a very elaborate process dependent upon accurate coordination of pre and post-synaptic specialization, requiring multiple steps and a variety of receptors and signaling molecules. Due to its relative structural simplicity and the ease in manipulation and observation, the neuromuscular synapse or neuromuscular junction (NMJ)-the connection between motor neurons and skeletal muscle-represents the archetype junction system for studying synapse formation and conservation. This junction is essential for survival, as it controls our ability to move and breath. NMJ formation requires coordinated interactions between motor neurons and muscle fibers, which ultimately result in the formation of a highly specialized post-synaptic architecture and a highly differentiated nerve terminal. Furthermore, to ensure a fast and reliable synaptic transmission following neurotransmitter release, ligand-gated channels (acetylcholine receptors, AChRs) are clustered on the post-synaptic muscle cell at high concentrations in sites opposite the presynaptic active zone, supporting a direct role for nerves in the organization of the post-synaptic membrane architecture. This organized clustering process, essential for NMJ formation and for life, relies on key signaling molecules and receptors and is regulated by soluble extracellular molecules localized within the synaptic cleft. Notably, several mutations as well as auto-antibodies against components of these signaling complexes have been related to neuromuscular disorders. The recent years have witnessed strong progress in the understanding of molecular identities, architectures, and functions of NMJ macromolecules. Among these, prominent roles have been proposed for neural variants of the proteoglycan agrin, its receptor at NMJs composed of the lipoprotein receptor-related protein 4 (LRP4) and the muscle-specific kinase (MuSK), as well as the regulatory soluble synapse-specific protease Neurotrypsin. In this review we summarize the current state of the art regarding molecular structures and (agrin-dependent) canonical, as well as (agrin-independent) non-canonical, MuSK signaling mechanisms that underscore the formation of neuromuscular junctions, with the aim of providing a broad perspective to further stimulate molecular, cellular and tissue biology investigations on this fundamental intercellular contact.
Collapse
Affiliation(s)
| | | | - Federico Forneris
- The Armenise-Harvard Laboratory of Structural Biology, Department Biology and Biotechnology, University of Pavia, Pavia, Italy
| |
Collapse
|
9
|
Canciani A, Catucci G, Forneris F. Structural characterization of the third scavenger receptor cysteine-rich domain of murine neurotrypsin. Protein Sci 2019; 28:746-755. [PMID: 30748049 DOI: 10.1002/pro.3587] [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: 10/03/2018] [Accepted: 02/11/2019] [Indexed: 01/05/2023]
Abstract
Neurotrypsin (NT) is a multi-domain serine protease of the nervous system with only one known substrate: the large proteoglycan Agrin. NT has seen to be involved in the maintenance/turnover of neuromuscular junctions and in processes of synaptic plasticity in the central nervous system. Roles which have been tied to its enzymatic activity, localized in the C-terminal serine-protease (SP) domain. However the purpose of NT's remaining 3-4 scavenger receptor cysteine-rich (SRCR) domains is still unclear. We have determined the crystal structure of the third SRCR domain of murine NT (mmNT-SRCR3), immediately preceding the SP domain and performed a comparative structural analysis using homologous SRCR structures. Our data and the elevated degree of structural conservation with homologous domains highlight possible functional roles for NT SRCRs. Computational and experimental analyses suggest the identification of a putative binding region for Ca2+ ions, known to regulate NT enzymatic activity. Furthermore, sequence and structure comparisons allow to single out regions of interest that, in future studies, might be implicated in Agrin recognition/binding or in interactions with as of yet undiscovered NT partners.
Collapse
Affiliation(s)
- Anselmo Canciani
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Via Ferrata 9/A, 27100 Pavia, Italy
| | - Gianluca Catucci
- Department of Life Sciences and Systems Biology, University of Torino, 10123 Turin, Italy
| | - Federico Forneris
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of Pavia, Via Ferrata 9/A, 27100 Pavia, Italy
| |
Collapse
|
10
|
Beta-Site Amyloid Precursor Protein Cleaving Enzyme 1 Inhibition Impairs Synaptic Plasticity via Seizure Protein 6. Biol Psychiatry 2018; 83:428-437. [PMID: 28129943 DOI: 10.1016/j.biopsych.2016.12.023] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 12/01/2016] [Accepted: 12/16/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) is a promising drug target for the treatment of Alzheimer's disease. Prolonged BACE1 inhibition interferes with structural and functional synaptic plasticity in mice, most likely by altering the metabolism of BACE1 substrates. Seizure protein 6 (SEZ6) is predominantly cleaved by BACE1, and Sez6 knockout mice share some phenotypes with BACE1 inhibitor-treated mice. We investigated whether SEZ6 is involved in BACE1 inhibition-induced structural and functional synaptic alterations. METHODS The function of NB-360, a novel blood-brain barrier penetrant and orally available BACE1 inhibitor, was verified by immunoblotting. In vivo microscopy was applied to monitor the impact of long-term pharmacological BACE1 inhibition on dendritic spines in the cerebral cortex of constitutive and conditional Sez6 knockout mice. Finally, synaptic functions were characterized using electrophysiological field recordings in hippocampal slices. RESULTS BACE1 enzymatic activity was strongly suppressed by NB-360. Prolonged NB-360 treatment caused a reversible spine density reduction in wild-type mice, but it did not affect Sez6-/- mice. Knocking out Sez6 in a small subset of mature neurons also prevented the structural postsynaptic changes induced by BACE1 inhibition. Hippocampal long-term potentiation was decreased in both chronic BACE1 inhibitor-treated wild-type mice and vehicle-treated Sez6-/- mice. However, chronic NB-360 treatment did not alter long-term potentiation in CA1 neurons of Sez6-/- mice. CONCLUSIONS Our results suggest that SEZ6 plays an important role in maintaining normal dendritic spine dynamics. Furthermore, SEZ6 is involved in BACE1 inhibition-induced structural and functional synaptic alterations.
Collapse
|
11
|
Henriques A, Croixmarie V, Bouscary A, Mosbach A, Keime C, Boursier-Neyret C, Walter B, Spedding M, Loeffler JP. Sphingolipid Metabolism Is Dysregulated at Transcriptomic and Metabolic Levels in the Spinal Cord of an Animal Model of Amyotrophic Lateral Sclerosis. Front Mol Neurosci 2018; 10:433. [PMID: 29354030 PMCID: PMC5758557 DOI: 10.3389/fnmol.2017.00433] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/15/2017] [Indexed: 12/11/2022] Open
Abstract
Lipid metabolism is drastically dysregulated in amyotrophic lateral sclerosis and impacts prognosis of patients. Animal models recapitulate alterations in the energy metabolism, including hypermetabolism and severe loss of adipose tissue. To gain insight into the molecular mechanisms underlying disease progression in amyotrophic lateral sclerosis, we have performed RNA-sequencing and lipidomic profiling in spinal cord of symptomatic SOD1G86R mice. Spinal transcriptome of SOD1G86R mice was characterized by differential expression of genes related to immune system, extracellular exosome, and lysosome. Hypothesis-driven identification of metabolites showed that lipids, including sphingomyelin(d18:0/26:1), ceramide(d18:1/22:0), and phosphatidylcholine(o-22:1/20:4) showed profound altered levels. A correlation between disease severity and gene expression or metabolite levels was found for sphingosine, ceramide(d18:1/26:0), Sgpp2, Sphk1, and Ugt8a. Joint-analysis revealed a significant enrichment of glycosphingolipid metabolism in SOD1G86R mice, due to the down-regulation of ceramide, glucosylceramide, and lactosylceramide and the overexpression of genes involved in their recycling in the lysosome. A drug-gene interaction database was interrogated to identify potential drugs able to modulate the dysregulated genes from the signaling pathway. Our results suggest that complex lipids are pivotally changed during the first phase of motor symptoms in an animal model of amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Alexandre Henriques
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, Strasbourg, France.,INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, Strasbourg, France.,Spedding Research Solutions SAS, Le Vesinet, France
| | | | - Alexandra Bouscary
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, Strasbourg, France.,INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, Strasbourg, France
| | - Althéa Mosbach
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, Strasbourg, France.,INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, Strasbourg, France
| | - Céline Keime
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM, U964, CNRS, UMR7104, Université de Strasbourg, Illkirch, France
| | | | | | | | - Jean-Philippe Loeffler
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, Strasbourg, France.,INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, Strasbourg, France
| |
Collapse
|
12
|
Hidaka C, Kashio T, Uchigaki D, Mitsui S. Vulnerability or resilience of motopsin knockout mice to maternal separation stress depending on adulthood behaviors. Neuropsychiatr Dis Treat 2018; 14:2255-2268. [PMID: 30233183 PMCID: PMC6129033 DOI: 10.2147/ndt.s170281] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Both environmental and genetic conditions contribute to the robust development of neuronal circuits and adulthood behaviors. Loss of motopsin gene function causes severe intellectual disability in humans and enhanced social behavior in mice. Furthermore, childhood maltreatment is a risk factor for some psychiatric disorders, and children with disabilities have a higher risk of abuse than healthy children. MATERIALS AND METHODS In this study, we investigated the effects of maternal separation (MS) on adulthood behaviors of motopsin knockout (KO) and wild-type (WT) mice. RESULTS The MS paradigm decreased the duration that WT mice stayed in the center area of an open field, but not for motopsin KO mice; however, it decreased the novel object recognition index in both genotypes. In the marble burying test, motopsin KO mice buried fewer marbles than WT mice, regardless of the rearing conditions. The MS paradigm slightly increased and reduced open arm entry in the elevated plus maze by WT and motopsin KO mice, respectively. In the three-chamber test, the rate of sniffing the animal cage was increased by the MS paradigm only for motopsin KO mice. After the three-chamber test, motopsin KO mice had fewer cFos-positive cells in the prelimbic cortex, which is involved in emotional response, than WT mice. In the infralimbic cortex, the MS paradigm decreased the number of cFos-positive cells in motopsin KO mice. CONCLUSION Our results suggest that motopsin deficiency and childhood adversity independently affect some behaviors, but they may interfere with each other for other behaviors. Defective neuronal circuits in the prefrontal cortex may add to this complexity.
Collapse
Affiliation(s)
- Chiharu Hidaka
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Japan, .,Department of Neurobiology and Anatomy, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Taiki Kashio
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Japan,
| | - Daiju Uchigaki
- Department of Occupational Therapy, Gunma University, Maebashi, Japan,
| | - Shinichi Mitsui
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Japan, .,Department of Occupational Therapy, Gunma University, Maebashi, Japan,
| |
Collapse
|
13
|
Selective phenylalanine to proline substitution for improved antimicrobial and anticancer activities of peptides designed on phenylalanine heptad repeat. Acta Biomater 2017; 57:170-186. [PMID: 28483698 DOI: 10.1016/j.actbio.2017.05.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/19/2017] [Accepted: 05/04/2017] [Indexed: 01/09/2023]
Abstract
Introducing cell-selectivity in antimicrobial peptides (AMPs) without compromising the antimicrobial and anti-endotoxin properties is a crucial step towards the development of new antimicrobial agents. A peptide designed on phenylalanine heptad repeat possesses significant cytotoxicity along with desired antimicrobial and anti-endotoxin properties. Amino acid substitutions at 'a' and/or 'd' positions of heptad repeats of AMPs could alter their helical structure in mammalian membrane-mimetic environments and cytotoxicity towards mammalian cells. Since proline is a helix breaker, effects of selective proline substitution(s) at 'a' and/or 'd' positions of a 15-residue peptide designed on phenylalanine heptad repeat (FR-15) were investigated. Proline-substituted FR-15 variants were highly selective toward bacteria and fungi over hRBCs and murine 3T3 cells and also retained their antibacterial activities at high salt, serum and elevated temperatures. These non-cytotoxic variants also inhibited LPS-induced production of pro-inflammatory cytokines/chemokines in human monocytes, THP-1, RAW 264.7 and in BALB/c mice. The two non-cytotoxic variants (FR8P and FR11P) showed potent anti-cancer activity against highly metastatic human breast cancer cell line MDA-MB-231 with IC50 values less than 10μM. At sub-IC50 concentrations, FR8P and FR11P also showed anti-migratory and anti-invasive effects against MDA-MB-231 cells. FR8P and FR11P induced cellular apoptosis by triggering intrinsic apoptotic pathway through depolarization of mitochondrial membrane potential and activation of caspases. Overall the results demonstrated the utilization of selective phenylalanine to proline substitution in a heptad repeat of phenylalanine residues for the design of cell-selective, broad-spectrum AMPs with significant anti-cancer properties. STATEMENT OF SIGNIFICANCE We have demonstrated a methodology to design cell-selective potent antimicrobial and anti-endotoxin peptides by utilizing phenylalanine zipper as a template and replacement of phenylalanine residue(s) from "a" and/or "d" position(s) with proline residue(s) produced non-cytotoxic AMPs with improved antibacterial properties against the drug-resistant strains of bacteria. The work showed that the 'a' and 'd' positions of the phenylalanine heptad repeat could be replaced by an appropriate amino acid to control cytotoxicity of the peptide without compromising its potency in antimicrobial and anti-endotoxin properties. The direct bacterial membrane targeting mechanism of proline substituted analogs of parent peptide makes difficult for bacteria to grow resistance against them. The peptides designed could be lead molecules in the area of sepsis as they possess significant anti-LPS activities for in vitro and in vivo. Interestingly since cancer cells and bacterial cell membranes possess the structural resemblances, the cancer cells are also targets for these peptides making them lead molecules in this field. However, unlike in bacteria where the peptides showed membrane permeabilization property to lyse them, the peptides induced apoptosis in MDA-MB-231 breast cancer cells to inhibit their proliferation and growth. The results are significant because it reveals that "a" and "d" positions of a phenylalanine zipper can be utilized as switches to design cell-selective, antimicrobial, anti-endotoxin and anticancer peptides.
Collapse
|
14
|
Piscidin-1-analogs with double L- and D-lysine residues exhibited different conformations in lipopolysaccharide but comparable anti-endotoxin activities. Sci Rep 2017; 7:39925. [PMID: 28051162 PMCID: PMC5209718 DOI: 10.1038/srep39925] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/10/2016] [Indexed: 01/11/2023] Open
Abstract
To become clinically effective, antimicrobial peptides (AMPs) should be non-cytotoxic to host cells. Piscidins are a group of fish-derived AMPs with potent antimicrobial and antiendotoxin activities but limited by extreme cytotoxicity. We conjectured that introduction of cationic residue(s) at the interface of polar and non-polar faces of piscidins may control their insertion into hydrophobic mammalian cell membrane and thereby reducing cytotoxicity. We have designed several novel analogs of piscidin-1 by substituting threonine residue(s) with L and D-lysine residue(s). L/D-lysine-substituted analogs showed significantly reduced cytotoxicity but exhibited either higher or comparable antibacterial activity akin to piscidin-1. Piscidin-1-analogs demonstrated higher efficacy than piscidin-1 in inhibiting lipopolysaccharide (LPS)-induced pro-inflammatory responses in THP-1 cells. T15,21K-piscidin-1 (0.5 mg/Kg) and T15,21dK-piscidin-1 (1.0 mg/Kg) demonstrated 100% survival of LPS (12.0 mg/Kg)-administered mice. High resolution NMR studies revealed that both piscidin-1 and T15,21K-piscidin-1 adopted helical structures, with latter showing a shorter helix, higher amphipathicity and cationic residues placed at optimal distances to form ionic/hydrogen bond with lipid A of LPS. Remarkably, T15,21dK-piscidin-1 showed a helix-loop-helix structure in LPS and its interactions with LPS could be sustained by the distance of separation of side chains of R7 and D-Lys-15 which is close to the inter-phosphate distance of lipid A.
Collapse
|
15
|
Functions of the Alzheimer's Disease Protease BACE1 at the Synapse in the Central Nervous System. J Mol Neurosci 2016; 60:305-315. [PMID: 27456313 PMCID: PMC5059407 DOI: 10.1007/s12031-016-0800-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 07/07/2016] [Indexed: 02/06/2023]
Abstract
Inhibition of the protease β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) is a promising treatment strategy for Alzheimer's disease, and a number of BACE inhibitors are currently progressing through clinical trials. The strategy aims to decrease production of amyloid-β (Aβ) peptide from the amyloid precursor protein (APP), thus reducing or preventing Aβ toxicity. Over the last decade, it has become clear that BACE1 proteolytically cleaves a number of substrates in addition to APP. These substrates are not known to be involved in the pathogenesis of Alzheimer's disease but have other roles in the developing and/or mature central nervous system. Consequently, BACE inhibition and knockout in mice results in synaptic and other neuronal dysfunctions and the key substrates responsible for these deficits are still being elucidated. Of the BACE1 substrates that have been validated to date, a number may contribute to the synaptic deficits seen with BACE blockade, including neuregulin 1, close homologue of L1 and seizure-related gene 6. It is important to understand the impact that BACE blockade may have on these substrates and other proteins detected in substrate screens and, if necessary, develop substrate-selective BACE inhibitors.
Collapse
|
16
|
Single Amino Acid Substitutions at Specific Positions of the Heptad Repeat Sequence of Piscidin-1 Yielded Novel Analogs That Show Low Cytotoxicity and In Vitro and In Vivo Antiendotoxin Activity. Antimicrob Agents Chemother 2016; 60:3687-99. [PMID: 27067326 DOI: 10.1128/aac.02341-15] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 03/27/2016] [Indexed: 12/16/2022] Open
Abstract
Piscidin-1 possesses significant antimicrobial and cytotoxic activities. To recognize the primary amino acid sequence(s) in piscidin-1 that could be important for its biological activity, a long heptad repeat sequence located in the region from amino acids 2 to 19 was identified. To comprehend the possible role of this motif, six analogs of piscidin-1 were designed by selectively replacing a single isoleucine residue at a d (5th) position or at an a (9th or 16th) position with either an alanine or a valine residue. Two more analogs, namely, I5F,F6A-piscidin-1 and V12I-piscidin-1, were designed for investigating the effect of interchanging an alanine residue at a d position with an adjacent phenylalanine residue and replacing a valine residue with an isoleucine residue at another d position of the heptad repeat of piscidin-1, respectively. Single alanine-substituted analogs exhibited significantly reduced cytotoxicity against mammalian cells compared with that of piscidin-1 but appreciably retained the antibacterial and antiendotoxin activities of piscidin-1. All the single valine-substituted piscidin-1 analogs and I5F,F6A-piscidin-1 showed cytotoxicity greater than that of the corresponding alanine-substituted analogs, antibacterial activity marginally greater than or similar to that of the corresponding alanine-substituted analogs, and also antiendotoxin activity superior to that of the corresponding alanine-substituted analogs. Interestingly, among these peptides, V12I-piscidin-1 showed the highest cytotoxicity and antibacterial and antiendotoxin activities. Lipopolysaccharide (12 mg/kg of body weight)-treated mice, further treated with I16A-piscidin-1, the piscidin-1 analog with the highest therapeutic index, at a single dose of 1 or 2 mg/kg of body weight, showed 80 and 100% survival, respectively. Structural and functional characterization of these peptides revealed the basis of their biological activity and demonstrated that nontoxic piscidin-1 analogs with significant antimicrobial and antiendotoxin activities can be designed by incorporating single alanine substitutions in the piscidin-1 heptad repeat.
Collapse
|
17
|
Hidaka C, Mitsui S. N-Glycosylation modulates filopodia-like protrusions induced by sez-6 through regulating the distribution of this protein on the cell surface. Biochem Biophys Res Commun 2015; 462:346-51. [PMID: 25960298 DOI: 10.1016/j.bbrc.2015.04.139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 04/29/2015] [Indexed: 11/26/2022]
Abstract
Seizure-related gene 6 (sez-6) is a trans-membrane protein expressed by neuronal cells that modulates dendritic branching. It has three clusters of eleven possible N-glycosylation sites in the extracellular domain region: sugar chain (SC)1-3, SC4-7, and SC8-11. Recent reports suggest that N-glycosylation modulates the membrane trafficking and function of trans-membrane proteins. Here, we studied the role of N-glycosylation in sez-6 function. We transfected mutants lacking one, two, or all N-glycosylation clusters into neuro2a cells. A mutant lacking all N-glycosylation was transported to the cell membrane. Mutants lacking one cluster (sez-6 ΔSC1-3, ΔSC4-7, ΔSC8-11) were evenly distributed on the cell membrane and secreted into the conditioned medium, as in wild-type sez-6; in contrast, the unglycosylated mutant, sez-6 ΔSC1-11, and mutants having only one cluster (sez-6 SC1-3, SC8-11) were localized in some portions on the cell membrane. Despite sez-6 SC4-7 having only one cluster, it was transported like the wild type. Among mutants behaving like the wild type, sez-6 ΔSC1-3 and ΔSC4-7 reduced neurite formation. Interestingly, mutants lacking SC4-7 (sez-6 ΔSC4-7) did not affect the formation of filopodia-like protrusions. In contrast, other mutants as well as the wild type induced it, suggesting that SC4-7 is crucial for filopodia-like protrusions. Our results indicate that N-glycosylation regulates cell morphology through modulating the cell surface distribution of sez-6 protein.
Collapse
Affiliation(s)
- Chiharu Hidaka
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514, Japan
| | - Shinichi Mitsui
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514, Japan.
| |
Collapse
|
18
|
Mitsui S, Osako Y, Yuri K. Mental retardation-related protease, motopsin (prss12), binds to the BRICHOS domain of the integral membrane protein 2a. Cell Biol Int 2013; 38:117-23. [PMID: 23955961 DOI: 10.1002/cbin.10164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 08/02/2013] [Indexed: 11/06/2022]
Abstract
Motopsin (prss12), a mosaic serine protease secreted by neuronal cells, is believed to be important for cognitive function, as the loss of its function causes severe nonsyndromic mental retardation. To understand the molecular role of motopsin, we identified the integral membrane protein 2a (Itm2a) as a motopsin-interacting protein using a yeast two-hybrid system. A pull-down assay showed that the BRICHOS domain of Itm2a was essential for this interaction. Motopsin and Itm2a co-localized in COS cells and in cultured neurons when transiently expressed in these cells. Both proteins were co-immunoprecipitated from lysates of these transfected COS cells. Itm2a was strongly detected in a brain lysate prepared between postnatal day 0 and 10, during which period motopsin protein was also enriched in the brain. Immunohistochemistry detected Itm2a as patchy spots along endothelial cells of brain capillaries (which also expressed myosin II regulatory light chain [RLC]), and on glial fibrillary acidic protein (GFAP)-positive processes in the developing cerebral cortex. The data raise the possibility that secreted motopsin interacts with endothelial cells in the developing brain.
Collapse
Affiliation(s)
- Shinichi Mitsui
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, Showa, Maebashi, 371-8514, Japan
| | | | | |
Collapse
|