1
|
Cui C, Xu B, Liu H, Wang C, Zhang T, Jiang P, Feng L. Exploring the Role of SMPD3 in the lncRNA-miRNA-mRNA Regulatory Network in TBI Progression by Influencing Energy Metabolism. J Inflamm Res 2024; 17:10835-10848. [PMID: 39677286 PMCID: PMC11646434 DOI: 10.2147/jir.s491290] [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: 10/07/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024] Open
Abstract
Background Traumatic brain injury (TBI) is associated with disturbances in energy metabolism. This study aimed to construct a lncRNA-miRNA-mRNA network through bioinformatics methods to explore energy metabolism-related genes in the pathogenesis of TBI. Methods Data from datasets GSE171718, GSE131695, and GSE223245 obtained from the Gene Expression Omnibus, were analyzed to identify differentially expressed (DE) genes. Regulatory relationships were investigated through miRDB, miRTarBase, and TargetScan, thereby forming a lncRNA-miRNA-mRNA network. The Molecular Signatures Database (MSigDB) was utilized to identify energy metabolism-related genes, and a protein-protein interaction (PPI) network was established through the STRING database. Functional annotation and enrichment analysis were conducted using GO and KEGG. The TBI mouse model was established to detect the expression levels of GOLGA8B, ZNF367, and SMPD3 in brain tissues. Results SMPD3 emerged as the key DE gene linked to energy metabolism in TBI, demonstrating a negative correlation with miR-218-5p and being associated with moderate unconsciousness and female patients. The PPI network revealed SMPD3 interactions with proteins associated with cell death, sphingolipid metabolism, and neurodegenerative diseases such as Alzheimer's disease. In vivo, GOLGA8B, ZNF367, and SMPD3 mRNA levels were significantly lower in TBI mice. Conclusion In summary, SMPD3 represents a crucial metabolic gene in the progression of TBI. It potentially provides a new therapeutic target for metabolic disorders caused by traumatic brain injury (TBI) and holds significant theoretical value for further research.
Collapse
Affiliation(s)
- Changmeng Cui
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Biao Xu
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Hui Liu
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Changshui Wang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Tao Zhang
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Lei Feng
- Department of Neurosurgery, Jining First People’s Hospital, Shandong First Medical University, Jining, Shandong, 272000, People’s Republic of China
| |
Collapse
|
2
|
Inskeep KA, Crase B, Dayarathna T, Stottmann RW. SMPD4-mediated sphingolipid metabolism regulates brain and primary cilia development. Development 2024; 151:dev202645. [PMID: 39470011 PMCID: PMC11586524 DOI: 10.1242/dev.202645] [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: 12/20/2023] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
Genetic variants in multiple sphingolipid biosynthesis genes cause human brain disorders. A recent study looked at people from 12 unrelated families with variants in the gene SMPD4, a neutral sphingomyelinase that metabolizes sphingomyelin into ceramide at an early stage of the biosynthesis pathway. These individuals have severe developmental brain malformations, including microcephaly and cerebellar hypoplasia. The disease mechanism of SMPD4 was not known and so we pursued a new mouse model. We hypothesized that the role of SMPD4 in producing ceramide is important for making primary cilia, a crucial organelle mediating cellular signaling. We found that the mouse model has cerebellar hypoplasia due to failure of Purkinje cell development. Human induced pluripotent stem cells lacking SMPD4 exhibit neural progenitor cell death and have shortened primary cilia, which is rescued by adding exogenous ceramide. SMPD4 production of ceramide is crucial for human brain development.
Collapse
Affiliation(s)
- Katherine A. Inskeep
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Bryan Crase
- Department of Neuroscience, The Ohio State University College of Arts and Sciences, Columbus, OH 43210, USA
| | - Thamara Dayarathna
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Rolf W. Stottmann
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| |
Collapse
|
3
|
Burgelman M, Dujardin P, Willems A, Hochepied T, Van Imschoot G, Van Wonterghem E, Van Hoecke L, Vandendriessche C, Vandenbroucke RE. Challenging the conventional wisdom: Re-evaluating Smpd3's role in extracellular vesicle biogenesis. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70015. [PMID: 39525277 PMCID: PMC11544639 DOI: 10.1002/jex2.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/27/2024] [Accepted: 09/28/2024] [Indexed: 11/16/2024]
Abstract
Extracellular vesicles (EVs) are pivotal in intercellular communication, impacting diverse physiological and pathological processes. Current in vitro EV biogenesis studies often utilize pharmacological inhibitors, inducing off-target effects and overlooking cell-specific production nuances. Addressing these limitations, we utilized CRISPR/Cas9 to generate heterozygous full-body and conditional sphingomyelin phosphodiesterase 3 (Smpd3) knockout (KO) transgenic mice. Smpd3, also known as neutral sphingomyelinase 2 (nSMase2), triggers membrane curvature through sphingomyelin hydrolysis to ceramide, thereby influencing exosome release. Intriguingly, Smpd3 deficiency demonstrated no impact on EV release both in vitro and in vivo, underscoring its potential cell-type-specific role in EV biogenesis. Notably, bone marrow derived macrophages (BMDMs) did exhibit reduced EV release upon Alix deletion. Our findings open avenues for subsequent inquiries, enriching our knowledge of EV biogenesis and illuminating intercellular communication in health and disease.
Collapse
Affiliation(s)
- Marlies Burgelman
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Pieter Dujardin
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Anthony Willems
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Tino Hochepied
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Griet Van Imschoot
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Elien Van Wonterghem
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Lien Van Hoecke
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Charysse Vandendriessche
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Roosmarijn E. Vandenbroucke
- VIB Center for Inflammation ResearchGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| |
Collapse
|
4
|
Zhu Z, McClintock TS, Bieberich E. Transcriptomics analysis reveals potential regulatory role of nSMase2 (Smpd3) in nervous system development and function of middle-aged mouse brains. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12911. [PMID: 39171374 PMCID: PMC11339599 DOI: 10.1111/gbb.12911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024]
Abstract
Neutral sphingomyelinase-2 (nSMase2), gene name sphingomyelin phosphodiesterase-3 (Smpd3), is a key regulatory enzyme responsible for generating the sphingolipid ceramide. The function of nSMase2 in the brain is still controversial. To better understand the functional roles of nSMase2 in the aging mouse brain, we applied RNA-seq analysis, which identified a total of 1462 differentially abundant mRNAs between +/fro and fro/fro, of which 891 were increased and 571 were decreased in nSMase2-deficient mouse brains. The most strongly enriched GO and KEGG annotation terms among transcripts increased in fro/fro mice included synaptogenesis, synapse development, synaptic signaling, axon development, and axonogenesis. Among decreased transcripts, enriched annotations included ribosome assembly and mitochondrial protein complex functions. KEGG analysis of decreased transcripts also revealed overrepresentation of annotations for Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington disease (HD). Ingenuity Pathway Analysis (IPA) tools predicted lower susceptibility to these neurodegenerative disorders, as well as predictions agreeing with stronger synaptic function, learning, and memory in fro/fro mice. The IPA tools identified signaling proteins, epigenetic regulators, and microRNAs as likely upstream regulators of the broader set of genes encoding the affected transcripts. It also revealed 16 gene networks, each linked to biological processes identified as overrepresented annotations among the affected transcripts by multiple analysis methods. Therefore, the analysis of these RNA-seq data indicates that nSMase2 impacts synaptic function and neural development, and may contribute to the onset and development of neurodegenerative diseases in middle-aged mice.
Collapse
Affiliation(s)
- Zhihui Zhu
- Department of PhysiologyUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Timothy S. McClintock
- Department of PhysiologyUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Erhard Bieberich
- Department of PhysiologyUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Veterans Affairs Medical CenterLexingtonKentuckyUSA
| |
Collapse
|
5
|
Doostparast Torshizi A, Truong DT, Hou L, Smets B, Whelan CD, Li S. Proteogenomic network analysis reveals dysregulated mechanisms and potential mediators in Parkinson's disease. Nat Commun 2024; 15:6430. [PMID: 39080267 PMCID: PMC11289099 DOI: 10.1038/s41467-024-50718-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
Parkinson's disease is highly heterogeneous across disease symptoms, clinical manifestations and progression trajectories, hampering the identification of therapeutic targets. Despite knowledge gleaned from genetics analysis, dysregulated proteome mechanisms stemming from genetic aberrations remain underexplored. In this study, we develop a three-phase system-level proteogenomic analytical framework to characterize disease-associated proteins and dysregulated mechanisms. Proteogenomic analysis identified 577 proteins that enrich for Parkinson's disease-related pathways, such as cytokine receptor interactions and lysosomal function. Converging lines of evidence identified nine proteins, including LGALS3, CSNK2A1, SMPD3, STX4, APOA2, PAFAH1B3, LDLR, HSPB1, BRK1, with potential roles in disease pathogenesis. This study leverages the largest population-scale proteomics dataset, the UK Biobank Pharma Proteomics Project, to characterize genetically-driven protein disturbances associated with Parkinson's disease. Taken together, our work contributes to better understanding of genome-proteome dynamics in Parkinson's disease and sets a paradigm to identify potential indirect mediators connected to GWAS signals for complex neurodegenerative disorders.
Collapse
Affiliation(s)
- Abolfazl Doostparast Torshizi
- Population Analytics & Insights, AI/ML, Data Science & Digital Health, Janssen Research & Development, LLC, Spring House, PA, USA.
| | - Dongnhu T Truong
- Population Analytics & Insights, AI/ML, Data Science & Digital Health, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Liping Hou
- Population Analytics & Insights, AI/ML, Data Science & Digital Health, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Bart Smets
- Neuroscience Data Science, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Christopher D Whelan
- Neuroscience Data Science, Janssen Research & Development, LLC, Cambridge, MA, USA
| | - Shuwei Li
- Population Analytics & Insights, AI/ML, Data Science & Digital Health, Janssen Research & Development, LLC, Spring House, PA, USA
| |
Collapse
|
6
|
Kim Y, Jeong M, Koh IG, Kim C, Lee H, Kim JH, Yurko R, Kim IB, Park J, Werling DM, Sanders SJ, An JY. CWAS-Plus: estimating category-wide association of rare noncoding variation from whole-genome sequencing data with cell-type-specific functional data. Brief Bioinform 2024; 25:bbae323. [PMID: 38966948 PMCID: PMC11224609 DOI: 10.1093/bib/bbae323] [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: 03/13/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/06/2024] Open
Abstract
Variants in cis-regulatory elements link the noncoding genome to human pathology; however, detailed analytic tools for understanding the association between cell-level brain pathology and noncoding variants are lacking. CWAS-Plus, adapted from a Python package for category-wide association testing (CWAS), enhances noncoding variant analysis by integrating both whole-genome sequencing (WGS) and user-provided functional data. With simplified parameter settings and an efficient multiple testing correction method, CWAS-Plus conducts the CWAS workflow 50 times faster than CWAS, making it more accessible and user-friendly for researchers. Here, we used a single-nuclei assay for transposase-accessible chromatin with sequencing to facilitate CWAS-guided noncoding variant analysis at cell-type-specific enhancers and promoters. Examining autism spectrum disorder WGS data (n = 7280), CWAS-Plus identified noncoding de novo variant associations in transcription factor binding sites within conserved loci. Independently, in Alzheimer's disease WGS data (n = 1087), CWAS-Plus detected rare noncoding variant associations in microglia-specific regulatory elements. These findings highlight CWAS-Plus's utility in genomic disorders and scalability for processing large-scale WGS data and in multiple-testing corrections. CWAS-Plus and its user manual are available at https://github.com/joonan-lab/cwas/ and https://cwas-plus.readthedocs.io/en/latest/, respectively.
Collapse
Affiliation(s)
- Yujin Kim
- Department of Integrated Biomedical and Life Science, Korea University, 145 Anam-ro, Seongbuk-ku, Seoul 02841, Republic of Korea
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, 145 Anam-ro, Seongbuk-ku, Seoul 02841, Republic of Korea
| | - Minwoo Jeong
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, 145 Anam-ro, Seongbuk-ku, Seoul 02841, Republic of Korea
| | - In Gyeong Koh
- Department of Integrated Biomedical and Life Science, Korea University, 145 Anam-ro, Seongbuk-ku, Seoul 02841, Republic of Korea
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, 145 Anam-ro, Seongbuk-ku, Seoul 02841, Republic of Korea
| | - Chanhee Kim
- Department of Integrated Biomedical and Life Science, Korea University, 145 Anam-ro, Seongbuk-ku, Seoul 02841, Republic of Korea
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, 145 Anam-ro, Seongbuk-ku, Seoul 02841, Republic of Korea
| | - Hyeji Lee
- Department of Integrated Biomedical and Life Science, Korea University, 145 Anam-ro, Seongbuk-ku, Seoul 02841, Republic of Korea
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, 145 Anam-ro, Seongbuk-ku, Seoul 02841, Republic of Korea
| | - Jae Hyun Kim
- Department of Integrated Biomedical and Life Science, Korea University, 145 Anam-ro, Seongbuk-ku, Seoul 02841, Republic of Korea
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, 145 Anam-ro, Seongbuk-ku, Seoul 02841, Republic of Korea
| | - Ronald Yurko
- Department of Statistics and Data Science, Carnegie Mellon University, 5000 Forbes Avenue, Squirrel Hill North, Pittsburgh, PA 15213, United States
| | - Il Bin Kim
- Department of Psychiatry, CHA Gangnam Medical Center, CHA University School of Medicine, 566 Nonhyon-ro, Gangnam-gu, Seoul 06135, Republic of Korea
| | - Jeongbin Park
- School of Biomedical Convergence Engineering, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do, 50612, Republic of Korea
| | - Donna M Werling
- Laboratory of Genetics, University of Wisconsin-Madison, 425-g Henry Mall, Madison, WI 53706, Unite States
| | - Stephan J Sanders
- Department of Paediatrics, Institute of Developmental and Regenerative Medicine, University of Oxford, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, United Kingdom
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, 1651 4th Street, San Francisco, CA 94158, United States
| | - Joon-Yong An
- Department of Integrated Biomedical and Life Science, Korea University, 145 Anam-ro, Seongbuk-ku, Seoul 02841, Republic of Korea
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, 145 Anam-ro, Seongbuk-ku, Seoul 02841, Republic of Korea
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, 145 Anam-ro, Seongbuk-ku, Seoul 02841, Republic of Korea
| |
Collapse
|
7
|
Piacentino ML, Fasse AJ, Camacho-Avila A, Grabylnikov I, Bronner ME. SMPD3 expression is spatially regulated in the developing embryo by SOXE factors. Dev Biol 2024; 506:31-41. [PMID: 38052296 PMCID: PMC10872304 DOI: 10.1016/j.ydbio.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/20/2023] [Accepted: 11/29/2023] [Indexed: 12/07/2023]
Abstract
During epithelial-to-mesenchymal transition (EMT), significant rearrangements occur in plasma membrane protein and lipid content that are important for membrane function and acquisition of cell motility. To gain insight into how neural crest cells regulate their lipid content at the transcriptional level during EMT, here we identify critical enhancer sequences that regulate the expression of SMPD3, a gene responsible for sphingomyelin hydrolysis to produce ceramide and necessary for neural crest EMT. We uncovered three enhancer regions within the first intron of the SMPD3 locus that drive reporter expression in distinct spatial and temporal domains, together collectively recapitulating the expression domains of endogenous SMPD3 within the ectodermal lineages. We further dissected one enhancer that is specifically active in the migrating neural crest. By mutating putative transcriptional input sites or knocking down upstream regulators, we find that the SOXE-family transcription factors SOX9 and SOX10 regulate the expression of SMPD3 in migrating neural crest cells. Further, ChIP-seq and nascent transcription analysis reveal that SOX10 directly regulates expression of an SMPD3 enhancer specific to migratory neural crest cells. Together these results shed light on how core components of developmental gene regulatory networks interact with metabolic effector genes to control changes in membrane lipid content.
Collapse
Affiliation(s)
- Michael L Piacentino
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Aria J Fasse
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Alexis Camacho-Avila
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Ilya Grabylnikov
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| |
Collapse
|
8
|
Inskeep KA, Crase B, Stottmann RW. SMPD4 mediated sphingolipid metabolism regulates brain and primary cilia development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.15.571873. [PMID: 38168190 PMCID: PMC10760124 DOI: 10.1101/2023.12.15.571873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Genetic variants in multiple sphingolipid biosynthesis genes cause human brain disorders. A recent study collected patients from twelve unrelated families with variants in the gene SMPD4 , a neutral sphingomyelinase which metabolizes sphingomyelin into ceramide at an early stage of the biosynthesis pathway. These patients have severe developmental brain malformations including microcephaly and cerebellar hypoplasia. However, the mechanism of SMPD4 was not known and we pursued a new mouse model. We hypothesized that the role of SMPD4 in producing ceramide is important for making primary cilia, a crucial organelle mediating cellular signaling. We found that the mouse model has cerebellar hypoplasia due to failure of Purkinje cell development. Human induced pluripotent stem cells exhibit neural progenitor cell death and have shortened primary cilia which is rescued by adding exogenous ceramide. SMPD4 production of ceramide is crucial for human brain development.
Collapse
|
9
|
Fernández-García P, Malet-Engra G, Torres M, Hanson D, Rosselló CA, Román R, Lladó V, Escribá PV. Evolving Diagnostic and Treatment Strategies for Pediatric CNS Tumors: The Impact of Lipid Metabolism. Biomedicines 2023; 11:biomedicines11051365. [PMID: 37239036 DOI: 10.3390/biomedicines11051365] [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: 03/10/2023] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Pediatric neurological tumors are a heterogeneous group of cancers, many of which carry a poor prognosis and lack a "standard of care" therapy. While they have similar anatomic locations, pediatric neurological tumors harbor specific molecular signatures that distinguish them from adult brain and other neurological cancers. Recent advances through the application of genetics and imaging tools have reshaped the molecular classification and treatment of pediatric neurological tumors, specifically considering the molecular alterations involved. A multidisciplinary effort is ongoing to develop new therapeutic strategies for these tumors, employing innovative and established approaches. Strikingly, there is increasing evidence that lipid metabolism is altered during the development of these types of tumors. Thus, in addition to targeted therapies focusing on classical oncogenes, new treatments are being developed based on a broad spectrum of strategies, ranging from vaccines to viral vectors, and melitherapy. This work reviews the current therapeutic landscape for pediatric brain tumors, considering new emerging treatments and ongoing clinical trials. In addition, the role of lipid metabolism in these neoplasms and its relevance for the development of novel therapies are discussed.
Collapse
Affiliation(s)
- Paula Fernández-García
- Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, 07122 Palma de Mallorca, Spain
- Laminar Pharmaceuticals, Isaac Newton, 07121 Palma de Mallorca, Spain
| | - Gema Malet-Engra
- Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, 07122 Palma de Mallorca, Spain
- Laminar Pharmaceuticals, Isaac Newton, 07121 Palma de Mallorca, Spain
| | - Manuel Torres
- Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, 07122 Palma de Mallorca, Spain
| | - Derek Hanson
- Hackensack Meridian Health, 343 Thornall Street, Edison, NJ 08837, USA
| | - Catalina A Rosselló
- Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, 07122 Palma de Mallorca, Spain
- Laminar Pharmaceuticals, Isaac Newton, 07121 Palma de Mallorca, Spain
| | - Ramón Román
- Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, 07122 Palma de Mallorca, Spain
- Laminar Pharmaceuticals, Isaac Newton, 07121 Palma de Mallorca, Spain
| | - Victoria Lladó
- Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, 07122 Palma de Mallorca, Spain
- Laminar Pharmaceuticals, Isaac Newton, 07121 Palma de Mallorca, Spain
| | - Pablo V Escribá
- Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, 07122 Palma de Mallorca, Spain
- Laminar Pharmaceuticals, Isaac Newton, 07121 Palma de Mallorca, Spain
| |
Collapse
|
10
|
Pal A, Gori S, Yoo SW, Thomas AG, Wu Y, Friedman J, Tenora L, Bhasin H, Alt J, Haughey N, Slusher BS, Rais R. Discovery of Orally Bioavailable and Brain-Penetrable Prodrugs of the Potent nSMase2 Inhibitor DPTIP. J Med Chem 2022; 65:11111-11125. [PMID: 35930706 PMCID: PMC9980655 DOI: 10.1021/acs.jmedchem.2c00562] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Extracellular vesicles (EVs) can carry pathological cargo and play an active role in disease progression. Neutral sphingomyelinase-2 (nSMase2) is a critical regulator of EV biogenesis, and its inhibition has shown protective effects in multiple disease states. 2,6-Dimethoxy-4-(5-phenyl-4-thiophen-2-yl-1H-imidazol-2-yl)phenol (DPTIP) is one of the most potent (IC50 = 30 nM) inhibitors of nSMase2 discovered to date. However, DPTIP exhibits poor oral pharmacokinetics (PK), limiting its clinical development. To overcome DPTIP's PK limitations, we synthesized a series of prodrugs by masking its phenolic hydroxyl group. When administered orally, the best prodrug (P18) with a 2',6'-diethyl-1,4'-bipiperidinyl promoiety exhibited >fourfold higher plasma (AUC0-t = 1047 pmol·h/mL) and brain exposures (AUC0-t = 247 pmol·h/g) versus DPTIP and a significant enhancement of DPTIP half-life (2 h vs ∼0.5 h). In a mouse model of acute brain injury, DPTIP released from P18 significantly inhibited IL-1β-induced EV release into plasma and attenuated nSMase2 activity. These studies report the discovery of a DPTIP prodrug with potential for clinical translation.
Collapse
Affiliation(s)
- Arindom Pal
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Sadakatali Gori
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Seung-wan Yoo
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Ajit G. Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Jacob Friedman
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Lukáš Tenora
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Harshit Bhasin
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Norman Haughey
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Barbara S. Slusher
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Department of Oncology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Departments of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Department of Medicine, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Corresponding Authors: . Tel: 410-502-0497. Fax: 410-614-0659 (R.R.), . Tel: 410-614-0662. Fax: 410-614-0659 (B.S.S.)
| | - Rana Rais
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Corresponding Authors: . Tel: 410-502-0497. Fax: 410-614-0659 (R.R.), . Tel: 410-614-0662. Fax: 410-614-0659 (B.S.S.)
| |
Collapse
|
11
|
Bost JP, Saher O, Hagey D, Mamand DR, Liang X, Zheng W, Corso G, Gustafsson O, Görgens A, Smith CIE, Zain R, El Andaloussi S, Gupta D. Growth Media Conditions Influence the Secretion Route and Release Levels of Engineered Extracellular Vesicles. Adv Healthc Mater 2022; 11:e2101658. [PMID: 34773385 PMCID: PMC11469210 DOI: 10.1002/adhm.202101658] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/01/2021] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) are nanosized cell-derived vesicles produced by all cells, which provide a route of intercellular communication by transmitting biological cargo. While EVs offer promise as therapeutic agents, the molecular mechanisms of EV biogenesis are not yet fully elucidated, in part due to the concurrence of numerous interwoven pathways which give rise to heterogenous EV populations in vitro. The equilibrium between the EV-producing pathways is heavily influenced by factors in the extracellular environment, in such a way that can be taken advantage of to boost production of engineered EVs. In this study, a quantifiable EV-engineering approach is used to investigate how different cell media conditions alter EV production. The presence of serum, exogenous EVs, and other signaling factors in cell media alters EV production at the physical, molecular, and transcriptional levels. Further, it is demonstrated that the ceramide-dependent EV biogenesis route is the major pathway to production of engineered EVs during optimized EV-production. These findings suggest a novel understanding to the mechanisms underlying EV production in cell culture which can be applied to develop advanced EV production methods.
Collapse
Affiliation(s)
- Jeremy P. Bost
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - Osama Saher
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
- Department of Pharmaceutics and Industrial PharmacyFaculty of PharmacyCairo UniversityCairo11562Egypt
| | - Daniel Hagey
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - Doste R. Mamand
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
- Department of BiologyFaculty of ScienceCihan University‐ErbilArbil5XC8+WVIraq
| | - Xiuming Liang
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - Wenyi Zheng
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - Giulia Corso
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - Oskar Gustafsson
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - André Görgens
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - CI Edvard Smith
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| | - Rula Zain
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
- Centre for Rare DiseasesDepartment of Clinical GeneticsKarolinska University HospitalStockholmSE‐171 76Sweden
| | | | - Dhanu Gupta
- Department of Laboratory MedicineKarolinska InstitutetHuddinge14152Sweden
| |
Collapse
|
12
|
Zeng P, Wang XM, Su HF, Zhang T, Ning LN, Shi Y, Yang SS, Lin L, Tian Q. Protective effects of Da-cheng-qi decoction in rats with intracerebral hemorrhage. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 90:153630. [PMID: 34217968 DOI: 10.1016/j.phymed.2021.153630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/15/2021] [Accepted: 06/10/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH), the most fatal subtype of stroke, has no disease-modifying treatment. Da-cheng-qi decoction (DCQ), composed of rhubarb, is one of the most commonly used Chinese traditional decoctions in ICH treatment. But the mechanism is not clear. Emodin is an active compound found in rhubarb. PURPOSE To study the protective effects of DCQ on ICH and its possible mechanisms of action. METHODS The ICH model was reproduced by injecting collagenase-VII into the left caudate putamen (CPu) of rats. DCQ and emodin were used to treat the ICH rats for 7 days. Behavior tests, proteomic analysis, morphological studies, and western blotting were performed. RESULTS The neurological deficits in the ICH rats recovered with DCQ and emodin on the 14th day after ICH. The proteomics data revealed that DCQ significantly corrected the pathological signals in the CPu and hippocampus after ICH. The numbers of amoebic microglia in the CPu and M2 microglia in both CPu and hippocampus were significantly increased after DCQ and emodin treatment. The increase in GluN2B-containing NMDA receptor (NR2B) and postsynaptic density protein-95, activation of mitogen-activated protein kinase (MAPK) signals in the CPu, and secondary neurodegeneration (SND) in the hippocampus were significantly recovered in DCQ-treated rats. Inhibition of MAPK p38 (p38) in the hippocampus was observed after DCQ and emodin treatment. CONCLUSION The protective effects of DCQ on ICH were confirmed in this study, and its mechanism may be related to the inhibition of MAPK and activation of M2 microglia. These results are beneficial to the development of ICH therapeutic targets.
Collapse
Affiliation(s)
- Peng Zeng
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Ming Wang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hong-Fei Su
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Teng Zhang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lin-Na Ning
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Shi
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shu-Sheng Yang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Traditional Chinese Medicine, Wuhan Red Cross Hospital, Wuhan 430015, China.
| | - Li Lin
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China; Laboratory of Medical Molecular and Cellular Biology, College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China.
| | - Qing Tian
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
13
|
Lecuyer M, Pathipati P, Faustino J, Vexler ZS. Neonatal stroke enhances interaction of microglia-derived extracellular vesicles with microglial cells. Neurobiol Dis 2021; 157:105431. [PMID: 34153465 PMCID: PMC9068249 DOI: 10.1016/j.nbd.2021.105431] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/21/2021] [Accepted: 06/16/2021] [Indexed: 12/15/2022] Open
Abstract
Microglial cells support brain homeostasis under physiological conditions and modulate brain injury in a context-dependent and brain maturation-dependent manner. Microglial cells protect neonatal brain from acute stroke. While microglial signaling via direct cell-cell interaction and release of variety of molecules is intensely studied, less is known about microglial signaling via release and uptake of extracellular vesicles (EVs). We asked whether neonatal stroke alters release of microglial EVs (MEV) and MEV communication with activated microglia. We pulled down and plated microglia from ischemic-reperfused and contralateral cortex 24 h after transient middle cerebral artery occlusion (tMCAO) in postnatal day 9 mice, isolated and characterized microglia-derived microvesicles (P3-MEV) and exosomes (P4-MEV), and determined uptake of fluorescently labeled P3-MEV and P4-MEV by plated microglia derived from ischemic-reperfused and contralateral cortex. We then examined how reducing EVs release in neonatal brain-by intra-cortical injection of CRISPR-Cas9-Smpd3/KO (Smpd3/KD) to downregulate Smpd3 gene to disrupt neutral sphingomyelinase-2 (N-SMase2)-impacts P3-MEV and P4-MEV release and stroke injury. Both size and protein composition differed between P3-MEV and P4-MEV. tMCAO further altered protein composition of P3-MEV and P4-MEV and significantly, up to 5-fold, increased uptake of both vesicle subtypes by microglia from ischemic-reperfused regions. Under physiological conditions neurons were the predominant cell type expressing N-SMase-2, an enzyme involved in lipid signaling and EVs release. After tMCAO N-SMase-2 expression was diminished in injured neurons but increased in activated microglia/macrophages, leading to overall reduced N-SMase-2 activity. Compared to intracerebral injection of control plasmid, CRISPR-Cas9-Smpd3/Ct, Smpd3/KD injection further reduced N-SMase-2 activity and significantly reduced injury. Smpd3 downregulation decreased MEV release from injured regions, reduced Smpd3/KD-P3-MEV uptake and abolished Smpd3/KD-P4-MEV uptake by microglia from ischemic-reperfused region. Cumulatively, these data demonstrate that microglial cells release both microvesicles and exosomes in naïve neonatal brain, that the state of microglial activation determines both properties of released EVs and their recognition/uptake by microglia in ischemic-reperfused and control regions, suggesting a modulatory role of MEV in neonatal stroke, and that sphingosine/N-SMase-2 signaling contributes both to EVs release and uptake (predominantly P4-MEV) after neonatal stroke.
Collapse
Affiliation(s)
| | | | - Joel Faustino
- Department of Neurology, UCSF, San Francisco, CA, USA
| | | |
Collapse
|
14
|
Florentinus-Mefailoski A, Bowden P, Scheltens P, Killestein J, Teunissen C, Marshall JG. The plasma peptides of Alzheimer's disease. Clin Proteomics 2021; 18:17. [PMID: 34182925 PMCID: PMC8240224 DOI: 10.1186/s12014-021-09320-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023] Open
Abstract
Background A practical strategy to discover proteins specific to Alzheimer’s dementia (AD) may be to compare the plasma peptides and proteins from patients with dementia to normal controls and patients with neurological conditions like multiple sclerosis or other diseases. The aim was a proof of principle for a method to discover proteins and/or peptides of plasma that show greater observation frequency and/or precursor intensity in AD. The endogenous tryptic peptides of Alzheimer’s were compared to normals, multiple sclerosis, ovarian cancer, breast cancer, female normal, sepsis, ICU Control, heart attack, along with their institution-matched controls, and normal samples collected directly onto ice. Methods Endogenous tryptic peptides were extracted from blinded, individual AD and control EDTA plasma samples in a step gradient of acetonitrile for random and independent sampling by LC–ESI–MS/MS with a set of robust and sensitive linear quadrupole ion traps. The MS/MS spectra were fit to fully tryptic peptides within proteins identified using the X!TANDEM algorithm. Observation frequency of the identified proteins was counted using SEQUEST algorithm. The proteins with apparently increased observation frequency in AD versus AD Control were revealed graphically and subsequently tested by Chi Square analysis. The proteins specific to AD plasma by Chi Square with FDR correction were analyzed by the STRING algorithm. The average protein or peptide log10 precursor intensity was compared across disease and control treatments by ANOVA in the R statistical system. Results Peptides and/or phosphopeptides of common plasma proteins such as complement C2, C7, and C1QBP among others showed increased observation frequency by Chi Square and/or precursor intensity in AD. Cellular gene symbols with large Chi Square values (χ2 ≥ 25, p ≤ 0.001) from tryptic peptides included KIF12, DISC1, OR8B12, ZC3H12A, TNF, TBC1D8B, GALNT3, EME2, CD1B, BAG1, CPSF2, MMP15, DNAJC2, PHACTR4, OR8B3, GCK, EXOSC7, HMGA1 and NT5C3A among others. Similarly, increased frequency of tryptic phosphopeptides were observed from MOK, SMIM19, NXNL1, SLC24A2, Nbla10317, AHRR, C10orf90, MAEA, SRSF8, TBATA, TNIK, UBE2G1, PDE4C, PCGF2, KIR3DP1, TJP2, CPNE8, and NGF amongst others. STRING analysis showed an increase in cytoplasmic proteins and proteins associated with alternate splicing, exocytosis of luminal proteins, and proteins involved in the regulation of the cell cycle, mitochondrial functions or metabolism and apoptosis. Increases in mean precursor intensity of peptides from common plasma proteins such as DISC1, EXOSC5, UBE2G1, SMIM19, NXNL1, PANO, EIF4G1, KIR3DP1, MED25, MGRN1, OR8B3, MGC24039, POLR1A, SYTL4, RNF111, IREB2, ANKMY2, SGKL, SLC25A5, CHMP3 among others were associated with AD. Tryptic peptides from the highly conserved C-terminus of DISC1 within the sequence MPGGGPQGAPAAAGGGGVSHRAGSRDCLPPAACFR and ARQCGLDSR showed a higher frequency and highest intensity in AD compared to all other disease and controls. Conclusion Proteins apparently expressed in the brain that were directly related to Alzheimer’s including Nerve Growth Factor (NFG), Sphingomyelin Phosphodiesterase, Disrupted in Schizophrenia 1 (DISC1), the cell death regulator retinitis pigmentosa (NXNl1) that governs the loss of nerve cells in the retina and the cell death regulator ZC3H12A showed much higher observation frequency in AD plasma vs the matched control. There was a striking agreement between the proteins known to be mutated or dis-regulated in the brains of AD patients with the proteins observed in the plasma of AD patients from endogenous peptides including NBN, BAG1, NOX1, PDCD5, SGK3, UBE2G1, SMPD3 neuronal proteins associated with synapse function such as KSYTL4, VTI1B and brain specific proteins such as TBATA. Supplementary Information The online version contains supplementary material available at 10.1186/s12014-021-09320-2.
Collapse
Affiliation(s)
- Angelique Florentinus-Mefailoski
- Ryerson Analytical Biochemistry Laboratory (RABL), Department of Chemistry and Biology, Faculty of Science, Ryerson University, 350 Victoria St., Toronto, ON, Canada
| | - Peter Bowden
- Ryerson Analytical Biochemistry Laboratory (RABL), Department of Chemistry and Biology, Faculty of Science, Ryerson University, 350 Victoria St., Toronto, ON, Canada
| | - Philip Scheltens
- Alzheimer Center, Dept of Neurology, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Joep Killestein
- MS Center, Dept of Neurology, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Charlotte Teunissen
- Neurochemistry Lab and Biobank, Dept of Clinical Chemistry, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - John G Marshall
- Ryerson Analytical Biochemistry Laboratory (RABL), Department of Chemistry and Biology, Faculty of Science, Ryerson University, 350 Victoria St., Toronto, ON, Canada. .,International Biobank of Luxembourg (IBBL), Luxembourg Institute of Health (Formerly CRP Sante Luxembourg), Strassen, Luxembourg.
| |
Collapse
|
15
|
Frankowska M, Jesus FM, Mühle C, Pacheco JV, Maior RS, Sadakierska‐Chudy A, Smaga I, Piechota M, Kalinichenko LS, Gulbins E, Kornhuber J, Filip M, Müller CP, Barros M. Cocaine attenuates acid sphingomyelinase activity during establishment of addiction-related behavior-A translational study in rats and monkeys. Addict Biol 2021; 26:e12955. [PMID: 32761719 DOI: 10.1111/adb.12955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/17/2020] [Accepted: 07/21/2020] [Indexed: 12/12/2022]
Abstract
Cocaine addiction is a severe psychiatric condition for which currently no effective pharmacotherapy is available. Brain mechanisms for the establishment of addiction-related behaviors are still not fully understood, and specific biomarkers for cocaine use are not available. Sphingolipids are major membrane lipids, which shape neuronal membrane composition and dynamics in the brain. Here, we investigated how chronic cocaine exposure during establishment of addiction-related behaviors affects the activity of the sphingolipid rheostat controlling enzymes in the brain of rats. As we detected specific effects on several enzymes in the brain, we tested whether the activity of selected enzymes in the blood may serve as potential biomarker for cocaine exposure in non-human primates (Callithrix penicillata). We found that intravenous cocaine self-administration led to a reduced mRNA expression of Cers1, Degs1 and Degs2, and Smpd1 in the prefrontal cortex of rats, as well as a reduction of Cers4 expression in the striatum. These effects reversed after 10 days of abstinence. Monkeys showed a robust cocaine-induced place preference (CPP). This coincided with a reduction in blood acid sphingomyelinase (ASM) activity after CPP establishment. This effect normalized after 15 days of abstinence. Altogether, these findings suggest that the establishment of cocaine addiction-related behaviors coincides with changes in the activity of sphingolipid controlling enzymes. In particular, blood ASM levels may serve as a translational biomarker for recent cocaine exposure.
Collapse
Affiliation(s)
- Małgorzata Frankowska
- Department of Drug Addiction Pharmacology, Polish Academy of Sciences Maj Institute of Pharmacology Krakow Poland
| | - Fernando M. Jesus
- Department of Pharmacy, School of Health Sciences University of Brasilia Brasilia Brazil
| | - Christiane Mühle
- Department of Psychiatry and Psychotherapy Friedrich‐Alexander‐University Erlangen‐Nürnberg (FAU) Erlangen Germany
| | - Jéssica V.N. Pacheco
- Department of Pharmacy, School of Health Sciences University of Brasilia Brasilia Brazil
| | - Rafael S. Maior
- Department of Physiological Sciences University of Brasília Brasilia Brazil
- Primate Center Institute of Biology, University of Brasilia Brasilia Brazil
| | - Anna Sadakierska‐Chudy
- Department of Drug Addiction Pharmacology, Polish Academy of Sciences Maj Institute of Pharmacology Krakow Poland
| | - Irena Smaga
- Department of Drug Addiction Pharmacology, Polish Academy of Sciences Maj Institute of Pharmacology Krakow Poland
| | - Marcin Piechota
- Department of Molecular Neuropharmacology, Polish Academy of Sciences Maj Institute of Pharmacology Krakow Poland
| | - Liubov S. Kalinichenko
- Department of Psychiatry and Psychotherapy Friedrich‐Alexander‐University Erlangen‐Nürnberg (FAU) Erlangen Germany
| | - Erich Gulbins
- Department of Molecular Biology University of Duisburg‐Essen Essen Germany
- Department of Surgery University of Cincinnati Cincinnati Ohio USA
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy Friedrich‐Alexander‐University Erlangen‐Nürnberg (FAU) Erlangen Germany
| | - Małgorzata Filip
- Department of Drug Addiction Pharmacology, Polish Academy of Sciences Maj Institute of Pharmacology Krakow Poland
| | - Christian P. Müller
- Department of Psychiatry and Psychotherapy Friedrich‐Alexander‐University Erlangen‐Nürnberg (FAU) Erlangen Germany
| | - Marilia Barros
- Department of Pharmacy, School of Health Sciences University of Brasilia Brasilia Brazil
- Primate Center Institute of Biology, University of Brasilia Brasilia Brazil
| |
Collapse
|
16
|
Neutral sphingomyelinase mediates the co-morbidity trias of alcohol abuse, major depression and bone defects. Mol Psychiatry 2021; 26:7403-7416. [PMID: 34584229 PMCID: PMC8872992 DOI: 10.1038/s41380-021-01304-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023]
Abstract
Mental disorders are highly comorbid and occur together with physical diseases, which are often considered to arise from separate pathogenic pathways. We observed in alcohol-dependent patients increased serum activity of neutral sphingomyelinase. A genetic association analysis in 456,693 volunteers found associations of haplotypes of SMPD3 coding for NSM-2 (NSM) with alcohol consumption, but also with affective state, and bone mineralisation. Functional analysis in mice showed that NSM controls alcohol consumption, affective behaviour, and their interaction by regulating hippocampal volume, cortical connectivity, and monoaminergic responses. Furthermore, NSM controlled bone-brain communication by enhancing osteocalcin signalling, which can independently supress alcohol consumption and reduce depressive behaviour. Altogether, we identified a single gene source for multiple pathways originating in the brain and bone, which interlink disorders of a mental-physical co-morbidity trias of alcohol abuse-depression/anxiety-bone disorder. Targeting NSM and osteocalcin signalling may, thus, provide a new systems approach in the treatment of a mental-physical co-morbidity trias.
Collapse
|
17
|
He Q, Liu H, Deng S, Chen X, Li D, Jiang X, Zeng W, Lu W. The Golgi Apparatus May Be a Potential Therapeutic Target for Apoptosis-Related Neurological Diseases. Front Cell Dev Biol 2020; 8:830. [PMID: 33015040 PMCID: PMC7493689 DOI: 10.3389/fcell.2020.00830] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/04/2020] [Indexed: 01/04/2023] Open
Abstract
Increasing evidence shows that, in addition to the classical function of protein processing and transport, the Golgi apparatus (GA) is also involved in apoptosis, one of the most common forms of cell death. The structure and the function of the GA is damaged during apoptosis. However, the specific effect of the GA on the apoptosis process is unclear; it may be involved in initiating or promoting apoptosis, or it may inhibit apoptosis. Golgi-related apoptosis is associated with a variety of neurological diseases including glioma, Alzheimer’s disease (AD), Parkinson’s disease (PD), and ischemic stroke. This review summarizes the changes and the possible mechanisms of Golgi structure and function during apoptosis. In addition, we also explore the possible mechanisms by which the GA regulates apoptosis and summarize the potential relationship between the Golgi and certain neurological diseases from the perspective of apoptosis. Elucidation of the interaction between the GA and apoptosis broadens our understanding of the pathological mechanisms of neurological diseases and provides new research directions for the treatment of these diseases. Therefore, we propose that the GA may be a potential therapeutic target for apoptosis-related neurological diseases.
Collapse
Affiliation(s)
- Qiang He
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hui Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Shuwen Deng
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiqian Chen
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Dong Li
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xuan Jiang
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Wenbo Zeng
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Wei Lu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
18
|
Bilousova T, Simmons BJ, Knapp RR, Elias CJ, Campagna J, Melnik M, Chandra S, Focht S, Zhu C, Vadivel K, Jagodzinska B, Cohn W, Spilman P, Gylys KH, Garg NK, John V. Dual Neutral Sphingomyelinase-2/Acetylcholinesterase Inhibitors for the Treatment of Alzheimer's Disease. ACS Chem Biol 2020; 15:1671-1684. [PMID: 32352753 PMCID: PMC8297715 DOI: 10.1021/acschembio.0c00311] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We report the discovery of a novel class of compounds that function as dual inhibitors of the enzymes neutral sphingomyelinase-2 (nSMase2) and acetylcholinesterase (AChE). Inhibition of these enzymes provides a unique strategy to suppress the propagation of tau pathology in the treatment of Alzheimer's disease (AD). We describe the key SAR elements that affect relative nSMase2 and/or AChE inhibitor effects and potency, in addition to the identification of two analogs that suppress the release of tau-bearing exosomes in vitro and in vivo. Identification of these novel dual nSMase2/AChE inhibitors represents a new therapeutic approach to AD and has the potential to lead to the development of truly disease-modifying therapeutics.
Collapse
Affiliation(s)
- Tina Bilousova
- Drug Discovery Laboratory, Department of Neurology, Mary S. Easton Center for Alzheimer's Disease Research, University of California, Los Angeles, California 90095, United States
- School of Nursing, University of California, Los Angeles, California 90095, United States
| | - Bryan J Simmons
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - Rachel R Knapp
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - Chris J Elias
- Drug Discovery Laboratory, Department of Neurology, Mary S. Easton Center for Alzheimer's Disease Research, University of California, Los Angeles, California 90095, United States
| | - Jesus Campagna
- Drug Discovery Laboratory, Department of Neurology, Mary S. Easton Center for Alzheimer's Disease Research, University of California, Los Angeles, California 90095, United States
| | - Mikhail Melnik
- School of Nursing, University of California, Los Angeles, California 90095, United States
| | - Sujyoti Chandra
- Drug Discovery Laboratory, Department of Neurology, Mary S. Easton Center for Alzheimer's Disease Research, University of California, Los Angeles, California 90095, United States
| | - Samantha Focht
- Drug Discovery Laboratory, Department of Neurology, Mary S. Easton Center for Alzheimer's Disease Research, University of California, Los Angeles, California 90095, United States
| | - Chunni Zhu
- Drug Discovery Laboratory, Department of Neurology, Mary S. Easton Center for Alzheimer's Disease Research, University of California, Los Angeles, California 90095, United States
| | - Kanagasabai Vadivel
- Drug Discovery Laboratory, Department of Neurology, Mary S. Easton Center for Alzheimer's Disease Research, University of California, Los Angeles, California 90095, United States
| | - Barbara Jagodzinska
- Drug Discovery Laboratory, Department of Neurology, Mary S. Easton Center for Alzheimer's Disease Research, University of California, Los Angeles, California 90095, United States
| | - Whitaker Cohn
- Drug Discovery Laboratory, Department of Neurology, Mary S. Easton Center for Alzheimer's Disease Research, University of California, Los Angeles, California 90095, United States
| | - Patricia Spilman
- Drug Discovery Laboratory, Department of Neurology, Mary S. Easton Center for Alzheimer's Disease Research, University of California, Los Angeles, California 90095, United States
| | - Karen H Gylys
- School of Nursing, University of California, Los Angeles, California 90095, United States
| | - Neil K Garg
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - Varghese John
- Drug Discovery Laboratory, Department of Neurology, Mary S. Easton Center for Alzheimer's Disease Research, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
19
|
Sen NE, Arsovic A, Meierhofer D, Brodesser S, Oberschmidt C, Canet-Pons J, Kaya ZE, Halbach MV, Gispert S, Sandhoff K, Auburger G. In Human and Mouse Spino-Cerebellar Tissue, Ataxin-2 Expansion Affects Ceramide-Sphingomyelin Metabolism. Int J Mol Sci 2019; 20:E5854. [PMID: 31766565 PMCID: PMC6928749 DOI: 10.3390/ijms20235854] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 02/08/2023] Open
Abstract
Ataxin-2 (human gene symbol ATXN2) acts during stress responses, modulating mRNA translation and nutrient metabolism. Ataxin-2 knockout mice exhibit progressive obesity, dyslipidemia, and insulin resistance. Conversely, the progressive ATXN2 gain of function due to the fact of polyglutamine (polyQ) expansions leads to a dominantly inherited neurodegenerative process named spinocerebellar ataxia type 2 (SCA2) with early adipose tissue loss and late muscle atrophy. We tried to understand lipid dysregulation in a SCA2 patient brain and in an authentic mouse model. Thin layer chromatography of a patient cerebellum was compared to the lipid metabolome of Atxn2-CAG100-Knockin (KIN) mouse spinocerebellar tissue. The human pathology caused deficits of sulfatide, galactosylceramide, cholesterol, C22/24-sphingomyelin, and gangliosides GM1a/GD1b despite quite normal levels of C18-sphingomyelin. Cerebellum and spinal cord from the KIN mouse showed a consistent decrease of various ceramides with a significant elevation of sphingosine in the more severely affected spinal cord. Deficiency of C24/26-sphingomyelins contrasted with excess C18/20-sphingomyelin. Spinocerebellar expression profiling revealed consistent reductions of CERS protein isoforms, Sptlc2 and Smpd3, but upregulation of Cers2 mRNA, as prominent anomalies in the ceramide-sphingosine metabolism. Reduction of Asah2 mRNA correlated to deficient S1P levels. In addition, downregulations for the elongase Elovl1, Elovl4, Elovl5 mRNAs and ELOVL4 protein explain the deficit of very long-chain sphingomyelin. Reduced ASMase protein levels correlated to the accumulation of long-chain sphingomyelin. Overall, a deficit of myelin lipids was prominent in SCA2 nervous tissue at prefinal stage and not compensated by transcriptional adaptation of several metabolic enzymes. Myelination is controlled by mTORC1 signals; thus, our human and murine observations are in agreement with the known role of ATXN2 yeast, nematode, and mouse orthologs as mTORC1 inhibitors and autophagy promoters.
Collapse
Affiliation(s)
- Nesli-Ece Sen
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
- Faculty of Biosciences, Goethe-University, 60438 Frankfurt am Main, Germany
| | - Aleksandar Arsovic
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - David Meierhofer
- Max Planck Institute for Molecular Genetics, Ihnestrasse 63-73, 14195 Berlin, Germany;
| | - Susanne Brodesser
- Membrane Biology and Lipid Biochemistry Unit, Life and Medical Sciences Institute, University of Bonn, 53121 Bonn, Germany;
| | - Carola Oberschmidt
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - Júlia Canet-Pons
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - Zeynep-Ece Kaya
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
- Cerrahpasa School of Medicine, Istanbul University, 34098 Istanbul, Turkey
| | - Melanie-Vanessa Halbach
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - Suzana Gispert
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - Konrad Sandhoff
- Membrane Biology and Lipid Biochemistry Unit, Life and Medical Sciences Institute, University of Bonn, 53121 Bonn, Germany;
| | - Georg Auburger
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| |
Collapse
|