1
|
Kitamura K, Saito K, Homma T, Fuyuki A, Onouchi S, Saito S. Prosaposin/Saposin Expression in the Developing Rat Olfactory and Vomeronasal Epithelia. J Dev Biol 2024; 12:29. [PMID: 39585030 PMCID: PMC11587001 DOI: 10.3390/jdb12040029] [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: 08/30/2024] [Revised: 10/15/2024] [Accepted: 11/04/2024] [Indexed: 11/26/2024] Open
Abstract
Prosaposin is a glycoprotein widely conserved in vertebrates, and it acts as a precursor for saposins that accelerate hydrolysis in lysosomes or acts as a neurotrophic factor without being processed into saposins. Neurogenesis in the olfactory neuroepithelia, including the olfactory epithelium (OE) and the vomeronasal epithelium (VNE), is known to occur throughout an animal's life, and mature olfactory neurons (ORNs) and vomeronasal receptor neurons (VRNs) have recently been revealed to express prosaposin in the adult olfactory organ. In this study, the expression of prosaposin in the rat olfactory organ during postnatal development was examined. In the OE, prosaposin immunoreactivity was observed in mature ORNs labeled using olfactory marker protein (OMP) from postnatal day (P) 0. Immature ORNs showed no prosaposin immunoreactivity throughout the examined period. In the VNE, OMP-positive VRNs were mainly observed in the basal region of the VNE on P10 and showed an adult-like distribution from P20. On the other hand, prosaposin immunoreactivity was observed in VRNs from P0, suggesting that not only mature VRNs but also immature VRNs express prosaposin. This study raises the possibility that prosaposin is required for the normal development of the olfactory organ and has different roles in the OE and the VNE.
Collapse
Affiliation(s)
- Kai Kitamura
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; (K.K.); (T.H.); (A.F.); (S.O.)
| | - Kyoko Saito
- Gifu Prefectural Chuo Livestock Hygiene Service Center, 1-1 Yanagido, Gifu 501-1112, Japan;
| | - Takeshi Homma
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; (K.K.); (T.H.); (A.F.); (S.O.)
| | - Aimi Fuyuki
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; (K.K.); (T.H.); (A.F.); (S.O.)
| | - Sawa Onouchi
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; (K.K.); (T.H.); (A.F.); (S.O.)
| | - Shouichiro Saito
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; (K.K.); (T.H.); (A.F.); (S.O.)
| |
Collapse
|
2
|
Yan L, Wen Z, Yang Y, Liu A, Li F, Zhang Y, Yang C, Li Y, Zhang Y. Dissecting the roles of prosaposin as an emerging therapeutic target for tumors and its underlying mechanisms. Biomed Pharmacother 2024; 180:117551. [PMID: 39405903 DOI: 10.1016/j.biopha.2024.117551] [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: 07/27/2024] [Revised: 09/22/2024] [Accepted: 10/08/2024] [Indexed: 11/14/2024] Open
Abstract
As a dual-function protein, prosaposin (PSAP) is a lysosome-associated protein that participates in a variety of cellular processes. In the lysosome, PSAP is processed to activate enzymes that degrade lipids. In addition, PSAP proteins located extracellularly are involved in cancer progression, such as proliferation and tumor death suppression signaling. Moreover, under different situations, PSAP exhibits distinct metastasis potentials in tumors. However, comprehensive insight into PSAP in cancer progression has been lacking. Here, we provide a framework of the role of PSAP in cancer and its clinical application in cancer patients, providing a novel perspective on the clinical translation of PSAP.
Collapse
Affiliation(s)
- Lirong Yan
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Zhenpeng Wen
- Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Yi Yang
- Department of Laboratory Animal Science, China Medical University, Shenyang, China
| | - Aoran Liu
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Fang Li
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Yuzhe Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Chunjiao Yang
- Department of Oncology, The Fifth Affiliated Hospital of Guangxi Medical University & The First People's Hospital of Nanning, Nanning, Guangxi, China
| | - Yanke Li
- Department of Anorectal Surgery, the First Hospital of China Medical University, Shenyang, China.
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China.
| |
Collapse
|
3
|
Taha HB, Birnbaum A, Matthews I, Aceituno K, Leon J, Thorwald M, Godoy-Lugo J, Cortes CJ. Activation of the muscle-to-brain axis ameliorates neurocognitive deficits in an Alzheimer's disease mouse model via enhancing neurotrophic and synaptic signaling. GeroScience 2024:10.1007/s11357-024-01345-3. [PMID: 39269584 DOI: 10.1007/s11357-024-01345-3] [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: 07/02/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Skeletal muscle regulates central nervous system (CNS) function and health, activating the muscle-to-brain axis through the secretion of skeletal muscle-originating factors ("myokines") with neuroprotective properties. However, the precise mechanisms underlying these benefits in the context of Alzheimer's disease (AD) remain poorly understood. To investigate muscle-to-brain axis signaling in response to amyloid β (Aβ)-induced toxicity, we generated 5xFAD transgenic female mice with enhanced skeletal muscle function (5xFAD;cTFEB;HSACre) at prodromal (4-months old) and late (8-months old) symptomatic stages. Skeletal muscle TFEB overexpression reduced Aβ plaque accumulation in the cortex and hippocampus at both ages and rescued behavioral neurocognitive deficits in 8-month-old 5xFAD mice. These changes were associated with transcriptional and protein remodeling of neurotrophic signaling and synaptic integrity, partially due to the CNS-targeting myokine prosaposin (PSAP). Our findings implicate the muscle-to-brain axis as a novel neuroprotective pathway against amyloid pathogenesis in AD.
Collapse
Affiliation(s)
- Hash Brown Taha
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90007, USA
| | - Allison Birnbaum
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Ian Matthews
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90007, USA
| | - Karel Aceituno
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90007, USA
| | - Jocelyne Leon
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90007, USA
| | - Max Thorwald
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90007, USA
| | - Jose Godoy-Lugo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90007, USA
| | - Constanza J Cortes
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90007, USA.
| |
Collapse
|
4
|
Wongdontri C, Luangtrakul W, Boonchuen P, Sarnow P, Somboonviwat K, Jaree P, Somboonwiwat K. Participation of shrimp pva-miR-166 in hemocyte homeostasis by modulating apoptosis-related gene PvProsaposin during white spot syndrome virus infection. J Virol 2024; 98:e0053024. [PMID: 39051786 PMCID: PMC11334483 DOI: 10.1128/jvi.00530-24] [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: 03/26/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Tiny controllers referred to as microRNAs (miRNAs) impede the expression of genes to modulate biological processes. In invertebrates, particularly in shrimp as a model organism, it has been demonstrated that miRNAs play a crucial role in modulating innate immune responses against viral infection. By analyzing small RNAs, we identified 60 differentially expressed miRNAs (DEMs) in Penaues vannamei hemocytes following infection with white spot syndrome virus (WSSV). We predicted the target genes of WSSV-responsive miRNAs, shedding light on their participation in diverse biological pathways. We are particularly intrigued by pva-miR-166, which is the most notably elevated miRNA among 60 DEMs. At 24 h post-infection (hpi), the negative correlation between the expression of pva-miR-166 and its target gene, PvProsaposin, was evident and their interaction was confirmed by a reduction in luciferase activity in vitro. Suppression of PvProsaposin in unchallenged shrimp led to decreased survival rates, reduced total hemocyte count (THC), and increased caspase 3/7 activity, suggesting its significant role in maintaining hemocyte homeostasis. In WSSV-infected shrimp, a lower number of hemocytes corresponded to a lower WSSV load, but higher shrimp mortality was observed when PvProsaposin was suppressed. Conformingly, the introduction of the pva-miR-166 mimic to WSSV-infected shrimp resulted in decreased levels of PvProsaposin transcripts, a significant loss of THC, and an increase in the hemocyte apoptosis. Taken together, we propose that pva-miR-166 modulates hemocyte homeostasis during WSSV infection by suppressing the PvProsaposin, an anti-apoptotic gene. PvProsaposin inhibition disrupts hemocyte homeostasis, rendering the shrimp's inability to withstand WSSV invasion.IMPORTANCEGene regulation by microRNAs (miRNAs) has been reported during viral infection. Furthermore, hemocytes serve a dual role, not only producing various immune-related molecules to combat viral infections but also acting as a viral replication site. Maintaining hemocyte homeostasis is pivotal for the shrimp's survival during infection. The upregulated miRNA pva-miR-166 could repress PvProsaposin expression in shrimp hemocytes infected with WSSV. The significance of PvProsaposin in maintaining hemocyte homeostasis via apoptosis led to reduced survival rate, decreased total hemocyte numbers, and elevated caspase 3/7 activity in PvProsaposin-silenced shrimp. Additionally, the inhibitory ability of pva-miR-166-mimic and dsRNA-PvProsaposin on the expression of PvProsaposin also lowered the THC, increases the hemocyte apoptosis, resulting in a lower WSSV copy number. Ultimately, the dysregulation of the anti-apoptotic gene PvProsaposin by pva-miR-166 during WSSV infection disrupts hemocyte homeostasis, leading to an immunocompromised state in shrimp, rendering them incapable of surviving WSSV invasion.
Collapse
Affiliation(s)
- Chantaka Wongdontri
- Department of Biochemistry, Center of Excellence for Molecular Biology and Genomics of Shrimp, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Waruntorn Luangtrakul
- Department of Biochemistry, Center of Excellence for Molecular Biology and Genomics of Shrimp, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Pakpoom Boonchuen
- School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Peter Sarnow
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Kulwadee Somboonviwat
- Department of Computer Engineering, Faculty of Engineering at Sriracha, Kasetsart University Sriracha Campus, Chonburi, Thailand
| | - Phattarunda Jaree
- Center of Applied Shrimp Research and Innovation, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Kunlaya Somboonwiwat
- Department of Biochemistry, Center of Excellence for Molecular Biology and Genomics of Shrimp, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
5
|
Franchini L, Porter JJ, Lueck JD, Orlandi C. Gz Enhanced Signal Transduction assaY (G ZESTY) for GPCR deorphanization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605282. [PMID: 39091869 PMCID: PMC11291178 DOI: 10.1101/2024.07.26.605282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
G protein-coupled receptors (GPCRs) are key pharmacological targets, yet many remain underutilized due to unknown activation mechanisms and ligands. Orphan GPCRs, lacking identified natural ligands, are a high priority for research, as identifying their ligands will aid in understanding their functions and potential as drug targets. Most GPCRs, including orphans, couple to Gi/o/z family members, however current assays to detect their activation are limited, hindering ligand identification efforts. We introduce GZESTY, a highly sensitive, cell-based assay developed in an easily deliverable format designed to study the pharmacology of Gi/o/z-coupled GPCRs and assist in deorphanization. We optimized assay conditions and developed an all-in-one vector employing novel cloning methods to ensure the correct expression ratio of GZESTY components. GZESTY successfully assessed activation of a library of ligand-activated GPCRs, detecting both full and partial agonism, as well as responses from endogenous GPCRs. Notably, with GZESTY we established the presence of endogenous ligands for GPR176 and GPR37 in brain extracts, validating its use in deorphanization efforts. This assay enhances the ability to find ligands for orphan GPCRs, expanding the toolkit for GPCR pharmacologists.
Collapse
Affiliation(s)
- Luca Franchini
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Joseph J. Porter
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - John D. Lueck
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Cesare Orlandi
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
6
|
Yin P, Wang H, Xue T, Yu X, Meng X, Mi Q, Song S, Xiong B, Bi Y, Yu L. Four-Dimensional Label-Free Quantitative Proteomics of Ginsenoside Rg 2 Ameliorated Scopolamine-Induced Memory Impairment in Mice through the Lysosomal Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14640-14652. [PMID: 38885433 DOI: 10.1021/acs.jafc.4c00181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease. Ginsenoside Rg2 has shown potential in treating AD, but the underlying protein regulatory mechanisms associated with ginsenoside Rg2 treatment for AD remain unclear. This study utilized scopolamine to induce memory impairment in mice, and proteomics methods were employed to investigate the potential molecular mechanism of ginsenoside Rg2 in treating AD model mice. The Morris water maze, hematoxylin and eosin staining, and Nissl staining results indicated that ginsenoside Rg2 enhanced cognitive ability and decreased neuronal damage in AD mice. Proteomics, western blot, and immunofluorescence results showed that ginsenoside Rg2 primarily improved AD mice by downregulating the expression of LGMN, LAMP1, and PSAP proteins through the regulation of the lysosomal pathway. Transmission electron microscopy and network pharmacology prediction results showed a potential connection between the mechanism of ginsenoside Rg2 treatment for AD mice and lysosomes. The comprehensive results indicated that ginsenoside Rg2 may improve AD by downregulating LGMN, LAMP1, and PSAP through the regulation of the lysosomal pathway.
Collapse
Affiliation(s)
- Pei Yin
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, People's Republic of China
| | - Heyu Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, People's Republic of China
| | - Tingfang Xue
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, People's Republic of China
| | - Xiaoran Yu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, People's Republic of China
| | - Xingjian Meng
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, People's Republic of China
| | - Qianwen Mi
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, People's Republic of China
| | - Shixin Song
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, People's Republic of China
| | - Boyu Xiong
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, People's Republic of China
| | - Yunfeng Bi
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, People's Republic of China
| | - Lei Yu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, People's Republic of China
| |
Collapse
|
7
|
Fuyuki A, Sohel MSH, Homma T, Kitamura K, Takashima S, Onouchi S, Saito S. Selective prosaposin expression in Langerhans islets of the mouse pancreas. Tissue Cell 2024; 88:102367. [PMID: 38537378 DOI: 10.1016/j.tice.2024.102367] [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: 12/18/2023] [Revised: 03/12/2024] [Accepted: 03/22/2024] [Indexed: 06/17/2024]
Abstract
The islets of Langerhans are clusters of endocrine cells surrounded by exocrine acinar cells in the pancreas. Prosaposin is a housekeeping protein required for normal lysosomal function, but its expression level is significantly different among tissues. Prosaposin also exists in various body fluids including serum. Intracellularly, prosaposin activates lysosomes and may support autophagy, and extracellularly, prosaposin promotes survival of neurons via G protein-coupled receptors. In this study, prosaposin and its mRNA expression were examined in endocrine cells of the islets as well as in exocrine acinar cells in the pancreas of mice by in situ hybridization and immunostaining. High expression levels of prosaposin were found in Alpha, Beta and Delta cells in the islets, whereas prosaposin mRNA expression was faint or negative and prosaposin immunoreactivity was negative in exocrine acinar cells. The high expression levels of prosaposin in endocrine cells may indicate that prosaposin plays a crucial role in crinophagy, which is a characteristic autophagy in peptide-secreting endocrine cells, and/or that prosaposin is secreted from pancreatic islets. Since prosaposin has been reported in serum, this study suggests a new possible function of the Langerhans islets.
Collapse
Affiliation(s)
- Aimi Fuyuki
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Md Shahriar Hasan Sohel
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Takeshi Homma
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Kai Kitamura
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Shigeo Takashima
- Division of Genomics Research, Life Science Research Center, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Sawa Onouchi
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Shouichiro Saito
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.
| |
Collapse
|
8
|
Holzapfel R, Prell A, Schumacher F, Perschin V, Friedmann Angeli JP, Kleuser B, Stigloher C, Fazeli G. Degradation of hexosylceramides is required for timely corpse clearance via formation of cargo-containing phagolysosomal vesicles. Eur J Cell Biol 2024; 103:151411. [PMID: 38582051 DOI: 10.1016/j.ejcb.2024.151411] [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: 08/24/2023] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/08/2024] Open
Abstract
Efficient degradation of phagocytic cargo in lysosomes is crucial to maintain cellular homeostasis and defending cells against pathogens. However, the mechanisms underlying the degradation and recycling of macromolecular cargo within the phagolysosome remain incompletely understood. We previously reported that the phagolysosome containing the corpse of the polar body in C. elegans tubulates into small vesicles to facilitate corpse clearance, a process that requires cargo protein degradation and amino acid export. Here we show that degradation of hexosylceramides by the prosaposin ortholog SPP-10 and glucosylceramidases is required for timely corpse clearance. We observed accumulation of membranous structures inside endolysosomes of spp-10-deficient worms, which are likely caused by increased hexosylceramide species. spp-10 deficiency also caused alteration of additional sphingolipid subclasses, like dihydroceramides, 2-OH-ceramides, and dihydrosphingomyelins. While corpse engulfment, initial breakdown of corpse membrane inside the phagolysosome and lumen acidification proceeded normally in spp-10-deficient worms, formation of the cargo-containing vesicles from the corpse phagolysosome was reduced, resulting in delayed cargo degradation and phagolysosome resolution. Thus, by combining ultrastructural studies and sphingolipidomic analysis with observing single phagolysosomes over time, we identified a role of prosaposin/SPP-10 in maintaining phagolysosomal structure, which promotes efficient resolution of phagocytic cargos.
Collapse
Affiliation(s)
- Rebecca Holzapfel
- Imaging Core Facility, Biocenter, University of Würzburg, Würzburg, Germany
| | - Agata Prell
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Fabian Schumacher
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany; Core-Facility BioSupraMol, Pharma-MS subunit, Freie Universität Berlin, Germany
| | - Veronika Perschin
- Imaging Core Facility, Biocenter, University of Würzburg, Würzburg, Germany
| | - José Pedro Friedmann Angeli
- Chair of Translational Cell Biology, Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Burkhard Kleuser
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | | | - Gholamreza Fazeli
- Chair of Translational Cell Biology, Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
9
|
Thulasiram MR, Yamamoto R, Olszewski RT, Gu S, Morell RJ, Hoa M, Dabdoub A. Molecular differences between neonatal and adult stria vascularis from organotypic explants and transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590986. [PMID: 38712156 PMCID: PMC11071502 DOI: 10.1101/2024.04.24.590986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Summary The stria vascularis (SV), part of the blood-labyrinth barrier, is an essential component of the inner ear that regulates the ionic environment required for hearing. SV degeneration disrupts cochlear homeostasis, leading to irreversible hearing loss, yet a comprehensive understanding of the SV, and consequently therapeutic availability for SV degeneration, is lacking. We developed a whole-tissue explant model from neonatal and adult mice to create a robust platform for SV research. We validated our model by demonstrating that the proliferative behaviour of the SV in vitro mimics SV in vivo, providing a representative model and advancing high-throughput SV research. We also provided evidence for pharmacological intervention in our system by investigating the role of Wnt/β-catenin signaling in SV proliferation. Finally, we performed single-cell RNA sequencing from in vivo neonatal and adult mouse SV and revealed key genes and pathways that may play a role in SV proliferation and maintenance. Together, our results contribute new insights into investigating biological solutions for SV-associated hearing loss. Significance Hearing loss impairs our ability to communicate with people and interact with our environment. This can lead to social isolation, depression, cognitive deficits, and dementia. Inner ear degeneration is a primary cause of hearing loss, and our study provides an in depth look at one of the major sites of inner ear degeneration: the stria vascularis. The stria vascularis and associated blood-labyrinth barrier maintain the functional integrity of the auditory system, yet it is relatively understudied. By developing a new in vitro model for the young and adult stria vascularis and using single cell RNA sequencing, our study provides a novel approach to studying this tissue, contributing new insights and widespread implications for auditory neuroscience and regenerative medicine. Highlights - We established an organotypic explant system of the neonatal and adult stria vascularis with an intact blood-labyrinth barrier. - Proliferation of the stria vascularis decreases with age in vitro , modelling its proliferative behaviour in vivo . - Pharmacological studies using our in vitro SV model open possibilities for testing injury paradigms and therapeutic interventions. - Inhibition of Wnt signalling decreases proliferation in neonatal stria vascularis.- We identified key genes and transcription factors unique to developing and mature SV cell types using single cell RNA sequencing.
Collapse
|
10
|
Labusch M, Thetiot M, Than-Trong E, Morizet D, Coolen M, Varet H, Legendre R, Ortica S, Mancini L, Bally-Cuif L. Prosaposin maintains adult neural stem cells in a state associated with deep quiescence. Stem Cell Reports 2024; 19:515-528. [PMID: 38518783 PMCID: PMC11096431 DOI: 10.1016/j.stemcr.2024.02.007] [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: 08/12/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/24/2024] Open
Abstract
In most vertebrates, adult neural stem cells (NSCs) continuously give rise to neurons in discrete brain regions. A critical process for maintaining NSC pools over long periods of time in the adult brain is NSC quiescence, a reversible and tightly regulated state of cell-cycle arrest. Recently, lysosomes were identified to regulate the NSC quiescence-proliferation balance. However, it remains controversial whether lysosomal activity promotes NSC proliferation or quiescence, and a finer influence of lysosomal activity on NSC quiescence duration or depth remains unexplored. Using RNA sequencing and pharmacological manipulations, we show that lysosomes are necessary for NSC quiescence maintenance. In addition, we reveal that expression of psap, encoding the lysosomal regulator Prosaposin, is enriched in quiescent NSCs (qNSCs) that reside upstream in the NSC lineage and display a deep/long quiescence phase in the adult zebrafish telencephalon. We show that shRNA-mediated psap knockdown increases the proportion of activated NSCs (aNSCs) as well as NSCs that reside in shallower quiescence states (signed by ascl1a and deltaA expression). Collectively, our results identify the lysosomal protein Psap as a (direct or indirect) quiescence regulator and unfold the interplay between lysosomal function and NSC quiescence heterogeneities.
Collapse
Affiliation(s)
- Miriam Labusch
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France; Sorbonne Université, Collège doctoral, 75005 Paris, France
| | - Melina Thetiot
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France
| | - Emmanuel Than-Trong
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France
| | - David Morizet
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France; Sorbonne Université, Collège doctoral, 75005 Paris, France
| | - Marion Coolen
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France
| | - Hugo Varet
- Institut Pasteur, Université Paris Cité, Platform Biomics, 75015 Paris, France
| | - Rachel Legendre
- Institut Pasteur, Université Paris Cité, Platform Biomics, 75015 Paris, France
| | - Sara Ortica
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France
| | - Laure Mancini
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France; Sorbonne Université, Collège doctoral, 75005 Paris, France
| | - Laure Bally-Cuif
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France.
| |
Collapse
|
11
|
Bang S, Jiang C, Xu J, Chandra S, McGinnis A, Luo X, He Q, Li Y, Wang Z, Ao X, Parisien M, Fernandes de Araujo LO, Jahangiri Esfahani S, Zhang Q, Tonello R, Berta T, Diatchenko L, Ji RR. Satellite glial GPR37L1 and its ligand maresin 1 regulate potassium channel signaling and pain homeostasis. J Clin Invest 2024; 134:e173537. [PMID: 38530364 PMCID: PMC11060744 DOI: 10.1172/jci173537] [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: 06/29/2023] [Accepted: 03/12/2024] [Indexed: 03/27/2024] Open
Abstract
G protein-coupled receptor 37-like 1 (GPR37L1) is an orphan GPCR with largely unknown functions. Here, we report that Gpr37l1/GRP37L1 ranks among the most highly expressed GPCR transcripts in mouse and human dorsal root ganglia (DRGs) and is selectively expressed in satellite glial cells (SGCs). Peripheral neuropathy induced by streptozotoxin (STZ) and paclitaxel (PTX) led to reduced GPR37L1 expression on the plasma membrane in mouse and human DRGs. Transgenic mice with Gpr37l1 deficiency exhibited impaired resolution of neuropathic pain symptoms following PTX- and STZ-induced pain, whereas overexpression of Gpr37l1 in mouse DRGs reversed pain. GPR37L1 is coexpressed with potassium channels, including KCNJ10 (Kir4.1) in mouse SGCs and both KCNJ3 (Kir3.1) and KCNJ10 in human SGCs. GPR37L1 regulates the surface expression and function of the potassium channels. Notably, the proresolving lipid mediator maresin 1 (MaR1) serves as a ligand of GPR37L1 and enhances KCNJ10- or KCNJ3-mediated potassium influx in SGCs through GPR37L1. Chemotherapy suppressed KCNJ10 expression and function in SGCs, which MaR1 rescued through GPR37L1. Finally, genetic analysis revealed that the GPR37L1-E296K variant increased chronic pain risk by destabilizing the protein and impairing the protein's function. Thus, GPR37L1 in SGCs offers a therapeutic target for the protection of neuropathy and chronic pain.
Collapse
Affiliation(s)
- Sangsu Bang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Changyu Jiang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Jing Xu
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Sharat Chandra
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Aidan McGinnis
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Xin Luo
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Qianru He
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Yize Li
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Zilong Wang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Xiang Ao
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Marc Parisien
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Lorenna Oliveira Fernandes de Araujo
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Sahel Jahangiri Esfahani
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Qin Zhang
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Raquel Tonello
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Luda Diatchenko
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurobiology and
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
12
|
Yu J, Li J, Matei N, Wang W, Tang L, Pang J, Li X, Fang L, Tang J, Zhang JH, Yan M. Intranasal administration of recombinant prosaposin attenuates neuronal apoptosis through GPR37/PI3K/Akt/ASK1 pathway in MCAO rats. Exp Neurol 2024; 373:114656. [PMID: 38114054 PMCID: PMC10922973 DOI: 10.1016/j.expneurol.2023.114656] [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: 09/27/2023] [Revised: 11/19/2023] [Accepted: 12/12/2023] [Indexed: 12/21/2023]
Abstract
Studies have reported that Prosaposin (PSAP) is neuroprotective in cerebrovascular diseases. We hypothesized that PSAP would reduce infarct volume by attenuating neuronal apoptosis and promoting cell survival through G protein-coupled receptor 37(GPR37)/PI3K/Akt/ASK1 pathway in middle cerebral artery occlusion (MCAO) rats. Two hundred and thirty-five male and eighteen female Sprague-Dawley rats were used. Recombinant human PSAP (rPSAP) was administered intranasally 1 h (h) after reperfusion. PSAP small interfering ribonucleic acid (siRNA), GPR37 siRNA, and PI3K specific inhibitor LY294002 were administered intracerebroventricularly 48 h before MCAO. Infarct volume, neurological score, immunofluorescence staining, Western blot, Fluoro-Jade C (FJC) and TUNEL staining were examined. The expression of endogenous PSAP and GPR37 were increased after MCAO. Intranasal administration of rPSAP reduced brain infarction, neuronal apoptosis, and improved both short- and long-term neurological function. Knockdown of endogenous PSAP aggravated neurological deficits. Treatment with exogenous rPSAP increased PI3K expression, Akt and ASK1 phosphorylation, and Bcl-2 expression; phosphorylated-JNK and Bax levels were reduced along with the number of FJC and TUNEL positive neurons. GPR37 siRNA and LY294002 abolished the anti-apoptotic effect of rPSAP at 24 h after MCAO. In conclusion, rPSAP attenuated neuronal apoptosis and improved neurological function through GPR37/PI3K/Akt/ASK1 pathway after MCAO in rats. Therefore, further exploration of PSAP as a potential treatment option in ischemic stroke is warranted.
Collapse
Affiliation(s)
- Jing Yu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Jinlan Li
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Nathanael Matei
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA; Department of Ophthalmology, University of Southern California, Los Angeles, CA 90007, USA
| | - Wenna Wang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Lihui Tang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Jinwei Pang
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Xue Li
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Lili Fang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiping Tang
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA.
| | - Min Yan
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
13
|
Gordon T. Brief Electrical Stimulation Promotes Recovery after Surgical Repair of Injured Peripheral Nerves. Int J Mol Sci 2024; 25:665. [PMID: 38203836 PMCID: PMC10779324 DOI: 10.3390/ijms25010665] [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: 10/13/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024] Open
Abstract
Injured peripheral nerves regenerate their axons in contrast to those in the central nervous system. Yet, functional recovery after surgical repair is often disappointing. The basis for poor recovery is progressive deterioration with time and distance of the growth capacity of the neurons that lose their contact with targets (chronic axotomy) and the growth support of the chronically denervated Schwann cells (SC) in the distal nerve stumps. Nonetheless, chronically denervated atrophic muscle retains the capacity for reinnervation. Declining electrical activity of motoneurons accompanies the progressive fall in axotomized neuronal and denervated SC expression of regeneration-associated-genes and declining regenerative success. Reduced motoneuronal activity is due to the withdrawal of synaptic contacts from the soma. Exogenous neurotrophic factors that promote nerve regeneration can replace the endogenous factors whose expression declines with time. But the profuse axonal outgrowth they provoke and the difficulties in their delivery hinder their efficacy. Brief (1 h) low-frequency (20 Hz) electrical stimulation (ES) proximal to the injury site promotes the expression of endogenous growth factors and, in turn, dramatically accelerates axon outgrowth and target reinnervation. The latter ES effect has been demonstrated in both rats and humans. A conditioning ES of intact nerve days prior to nerve injury increases axonal outgrowth and regeneration rate. Thereby, this form of ES is amenable for nerve transfer surgeries and end-to-side neurorrhaphies. However, additional surgery for applying the required electrodes may be a hurdle. ES is applicable in all surgeries with excellent outcomes.
Collapse
Affiliation(s)
- Tessa Gordon
- Division of Reconstructive Surgery, Department of Surgery, University of Toronto, Toronto, ON M4G 1X8, Canada
| |
Collapse
|
14
|
Bang S, Jiang C, Xu J, Chandra S, McGinnis A, Luo X, He Q, Li Y, Wang Z, Ao X, Parisien M, Fernandes de Araujo LO, Esfahan SJ, Zhang Q, Tonello R, Berta T, Diatchenko L, Ji RR. Satellite glial GPR37L1 regulates maresin and potassium channel signaling for pain control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.03.569787. [PMID: 38106084 PMCID: PMC10723316 DOI: 10.1101/2023.12.03.569787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
G protein coupled receptor 37-like 1 (GPR37L1) is an orphan GPCR and its function remains largely unknown. Here we report that GPR37L1 transcript is highly expressed compared to all known GPCRs in mouse and human dorsal root ganglia (DRGs) and selectively expressed in satellite glial cells (SGCs). Peripheral neuropathy following diabetes and chemotherapy by streptozotocin and paclitaxel resulted in downregulations of surface GPR37L1 in mouse and human DRGs. Transgenic mice with Gpr37l1 deficiency exhibited impaired resolution of neuropathic pain symptom (mechanical allodynia), whereas overexpression of Gpr37l1 in mouse DRGs can reverse neuropathic pain. Notably, GPR37L1 is co-expressed and coupled with potassium channels in SGCs. We found striking species differences in potassium channel expression in SGCs, with predominant expression of KCNJ10 and KCNJ3 in mouse and human SGCs, respectively. GPR37L1 regulates the surface expression and function of KCNJ10 and KCNJ3. We identified the pro-resolving lipid mediator maresin 1 (MaR1) as a GPR37L1 ligand. MaR1 increases KCNJ10/KCNJ3-mediated potassium influx in SGCs via GPR37L1. MaR1 protected chemotherapy-induced suppression of KCNJ13/KCNJ10 expression and function in SGCs. Finally, genetic analysis revealed that the GPR37L1-E296K variant is associated with increased chronic pain risk by destabilizing the protein. Thus, GPR37L1 in SGCs offers a new target for neuropathy protection and pain control.
Collapse
|
15
|
Qiu X, Guo Y, Liu M, Zhang B, Li J, Wei J, Li M. Single-cell RNA-sequencing analysis reveals enhanced non-canonical neurotrophic factor signaling in the subacute phase of traumatic brain injury. CNS Neurosci Ther 2023; 29:3446-3459. [PMID: 37269057 PMCID: PMC10580338 DOI: 10.1111/cns.14278] [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: 02/09/2023] [Revised: 04/25/2023] [Accepted: 05/14/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a leading cause of long-term disability in young adults and induces complex neuropathological processes. Cellular autonomous and intercellular changes during the subacute phase contribute substantially to the neuropathology of TBI. However, the underlying mechanisms remain elusive. In this study, we explored the dysregulated cellular signaling during the subacute phase of TBI. METHODS Single-cell RNA-sequencing data (GSE160763) of TBI were analyzed to explore the cell-cell communication in the subacute phase of TBI. Upregulated neurotrophic factor signaling was validated in a mouse model of TBI. Primary cell cultures and cell lines were used as in vitro models to examine the potential mechanisms affecting signaling. RESULTS Single-cell RNA-sequencing analysis revealed that microglia and astrocytes were the most affected cells during the subacute phase of TBI. Cell-cell communication analysis demonstrated that signaling mediated by the non-canonical neurotrophic factors midkine (MDK), pleiotrophin (PTN), and prosaposin (PSAP) in the microglia/astrocytes was upregulated in the subacute phase of TBI. Time-course profiling showed that MDK, PTN, and PSAP expression was primarily upregulated in the subacute phase of TBI, and astrocytes were the major source of MDK and PTN after TBI. In vitro studies revealed that the expression of MDK, PTN, and PSAP in astrocytes was enhanced by activated microglia. Moreover, MDK and PTN promoted the proliferation of neural progenitors derived from human-induced pluripotent stem cells (iPSCs) and neurite growth in iPSC-derived neurons, whereas PSAP exclusively stimulated neurite growth. CONCLUSION The non-canonical neurotrophic factors MDK, PTN, and PSAP were upregulated in the subacute phase of TBI and played a crucial role in neuroregeneration.
Collapse
Affiliation(s)
- Xuecheng Qiu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouJiangsuChina
| | - Yaling Guo
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouJiangsuChina
| | - Ming‐Feng Liu
- Department of NeurosurgeryXuzhou Hospital of Traditional Chinese MedicineXuzhouJiangsuChina
| | - Bingge Zhang
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouJiangsuChina
| | - Jingzhen Li
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouJiangsuChina
| | - Jian‐Feng Wei
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouJiangsuChina
- Department of Histology and EmbryologyXuzhou Medical UniversityXuzhouJiangsuChina
| | - Meng Li
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouJiangsuChina
| |
Collapse
|
16
|
Yamamiya K, Li X, Nabeka H, Khan S, Khan F, Wakisaka H, Saito S, Hamada F, Matsuda S. Tracking of Prosaposin, a Saposin Precursor, in Rat Testis. J Histochem Cytochem 2023; 71:537-554. [PMID: 37728096 PMCID: PMC10546980 DOI: 10.1369/00221554231198570] [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: 03/27/2023] [Accepted: 08/02/2023] [Indexed: 09/21/2023] Open
Abstract
We tracked prosaposin (PSAP), a trophic factor, using an antibody specific to its proteolytic portion and an antibody to sortilin that traffics PSAP only to the lysosome. Immunostaining revealed that PSAP was distributed mainly on the basal side of seminiferous tubules, where many Sertoli cells and pachytene spermatocytes contained PSAP and its distribution differed depending on the stage of the spermatogenic cycle. The PSAP-sortilin complex was sorted to large lysosomes in the basal cytoplasm of Sertoli cells, where it may be processed into saposins. In contrast, in the thinner apical cytoplasm of Sertoli cells, PSAP in small lysosomes was transported to the apical side around sperm heads or into the lumen for secretion. The results of in situ hybridization analyses suggested that immature tubular cells in young animals produce PSAP to self-stimulate proliferation. However, in adults, not only Sertoli cells but also pachytene spermatocytes produce and secrete PSAP around germ cells or into the tubular lumen to stimulate cell proliferation or differentiation in a paracrine or autocrine manner. In summary, PSAP is not only a precursor of lysosomal enzymes but also a pivotal trophic factor in organogenesis in the immature testis and spermatogenesis in the mature testis.
Collapse
Affiliation(s)
| | - Xuan Li
- Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Bolinger AA, Frazier A, La JH, Allen JA, Zhou J. Orphan G Protein-Coupled Receptor GPR37 as an Emerging Therapeutic Target. ACS Chem Neurosci 2023; 14:3318-3334. [PMID: 37676000 PMCID: PMC11144446 DOI: 10.1021/acschemneuro.3c00479] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are successful druggable targets, making up around 35% of all FDA-approved medications. However, a large number of receptors remain orphaned, with no known endogenous ligand, representing a challenging but untapped area to discover new therapeutic targets. Among orphan GPCRs (oGPCRs) of interest, G protein-coupled receptor 37 (GPR37) is highly expressed in the central nervous system (CNS), particularly in the spinal cord and oligodendrocytes. While its cellular signaling mechanisms and endogenous receptor ligands remain elusive, GPR37 has been implicated in several important neurological conditions, including Parkinson's disease (PD), inflammation, pain, autism, and brain tumors. GPR37 structure, signaling, emerging physiology, and pharmacology are reviewed while integrating a discussion on potential therapeutic indications and opportunities.
Collapse
Affiliation(s)
- Andrew A. Bolinger
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Andrew Frazier
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jun-Ho La
- Department of Neurobiology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - John A. Allen
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jia Zhou
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
18
|
He Y, Kaya I, Shariatgorji R, Lundkvist J, Wahlberg LU, Nilsson A, Mamula D, Kehr J, Zareba-Paslawska J, Biverstål H, Chergui K, Zhang X, Andren PE, Svenningsson P. Prosaposin maintains lipid homeostasis in dopamine neurons and counteracts experimental parkinsonism in rodents. Nat Commun 2023; 14:5804. [PMID: 37726325 PMCID: PMC10509278 DOI: 10.1038/s41467-023-41539-5] [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: 05/08/2022] [Accepted: 09/08/2023] [Indexed: 09/21/2023] Open
Abstract
Prosaposin (PSAP) modulates glycosphingolipid metabolism and variants have been linked to Parkinson's disease (PD). Here, we find altered PSAP levels in the plasma, CSF and post-mortem brain of PD patients. Altered plasma and CSF PSAP levels correlate with PD-related motor impairments. Dopaminergic PSAP-deficient (cPSAPDAT) mice display hypolocomotion and depression/anxiety-like symptoms with mildly impaired dopaminergic neurotransmission, while serotonergic PSAP-deficient (cPSAPSERT) mice behave normally. Spatial lipidomics revealed an accumulation of highly unsaturated and shortened lipids and reduction of sphingolipids throughout the brains of cPSAPDAT mice. The overexpression of α-synuclein via AAV lead to more severe dopaminergic degeneration and higher p-Ser129 α-synuclein levels in cPSAPDAT mice compared to WT mice. Overexpression of PSAP via AAV and encapsulated cell biodelivery protected against 6-OHDA and α-synuclein toxicity in wild-type rodents. Thus, these findings suggest PSAP may maintain dopaminergic lipid homeostasis, which is dysregulated in PD, and counteract experimental parkinsonism.
Collapse
Affiliation(s)
- Yachao He
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Ibrahim Kaya
- Department of Pharmaceutical Biosciences, Medical Mass Spectrometry Imaging, Uppsala University, Uppsala, Sweden
| | - Reza Shariatgorji
- Department of Pharmaceutical Biosciences, Medical Mass Spectrometry Imaging, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Spatial Mass Spectrometry, Uppsala University, Uppsala, Sweden
| | - Johan Lundkvist
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden
- Sinfonia Biotherapeutics AB, Huddinge, Sweden
| | - Lars U Wahlberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden
| | - Anna Nilsson
- Department of Pharmaceutical Biosciences, Medical Mass Spectrometry Imaging, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Spatial Mass Spectrometry, Uppsala University, Uppsala, Sweden
| | - Dejan Mamula
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jan Kehr
- Section of Pharmacological Neurochemistry, Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Justyna Zareba-Paslawska
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Biverstål
- Sinfonia Biotherapeutics AB, Huddinge, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Karima Chergui
- Laboratory of Molecular Neurophysiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Xiaoqun Zhang
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Per E Andren
- Department of Pharmaceutical Biosciences, Medical Mass Spectrometry Imaging, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Spatial Mass Spectrometry, Uppsala University, Uppsala, Sweden
| | - Per Svenningsson
- Translational Neuropharmacology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
19
|
Bhattacharya P, Dhawan UK, Hussain MT, Singh P, Bhagat KK, Singhal A, Austin-Williams S, Sengupta S, Subramanian M. Efferocytes release extracellular vesicles to resolve inflammation and tissue injury via prosaposin-GPR37 signaling. Cell Rep 2023; 42:112808. [PMID: 37436891 DOI: 10.1016/j.celrep.2023.112808] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/16/2023] [Accepted: 06/27/2023] [Indexed: 07/14/2023] Open
Abstract
Macrophages release soluble mediators following efferocytic clearance of apoptotic cells to facilitate intercellular communication and promote the resolution of inflammation. However, whether inflammation resolution is modulated by extracellular vesicles (EVs) and vesicular mediators released by efferocytes is not known. We report that efferocyte-derived EVs express prosaposin, which binds to macrophage GPR37 to increase expression of the efferocytosis receptor Tim4 via an ERK-AP1-dependent signaling axis, leading to increased macrophage efferocytosis efficiency and accelerated resolution of inflammation. Neutralization and knockdown of prosaposin or blocking GRP37 abrogates the pro-resolution effects of efferocyte-derived EVs in vivo. Administration of efferocyte-derived EVs in a murine model of atherosclerosis is associated with an increase in lesional macrophage efferocytosis efficiency and a decrease in plaque necrosis and lesional inflammation. Thus, we establish a critical role for efferocyte-derived vesicular mediators in increasing macrophage efferocytosis efficiency and accelerating the resolution of inflammation and tissue injury.
Collapse
Affiliation(s)
- Purbasha Bhattacharya
- CSIR - Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Umesh Kumar Dhawan
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mohammed Tayab Hussain
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Praveen Singh
- CSIR - Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Karran Kiran Bhagat
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Aarushi Singhal
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Shani Austin-Williams
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Shantanu Sengupta
- CSIR - Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Manikandan Subramanian
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
20
|
Sohel MSH, Atoji Y, Onouchi S, Saito S. Expression patterns of prosaposin and neurotransmitter-related molecules in the chick paratympanic organ. Tissue Cell 2023; 83:102130. [PMID: 37320868 DOI: 10.1016/j.tice.2023.102130] [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: 05/02/2023] [Revised: 06/01/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023]
Abstract
The paratympanic organ (PTO) is a small sense organ in the middle ear of birds that contains hair cells similar to those found in vestibuloauditory organs and receives afferent fibers from the geniculate ganglion. To consider the histochemical similarities between the PTO and vestibular hair cells, we examined the expression patterns of representative molecules in vestibular hair cells, including prosaposin, G protein-coupled receptor (GPR) 37 and GPR37L1 as prosaposin receptors, vesicular glutamate transporter (vGluT) 2 and vGluT3, nicotinic acetylcholine receptor subunit α9 (nAChRα9), and glutamic acid decarboxylase (GAD) 65 and GAD67, in the postnatal day 0 chick PTO and geniculate ganglion by in situ hybridization. Prosaposin mRNA was observed in PTO hair cells, supporting cells, and geniculate ganglion cells. vGluT3 mRNA was observed in PTO hair cells, whereas vGluT2 was observed in a small number of ganglion cells. nAChRα9 mRNA was observed in a small number of PTO hair cells. The results suggest that the histochemical character of PTO hair cells is more similar to that of vestibular hair cells than that of auditory hair cells in chicks.
Collapse
Affiliation(s)
- Md Shahriar Hasan Sohel
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Yasuro Atoji
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Sawa Onouchi
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Shouichiro Saito
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.
| |
Collapse
|
21
|
Lee JM, Choi YJ, Yoo MC, Yeo SG. Central Facial Nervous System Biomolecules Involved in Peripheral Facial Nerve Injury Responses and Potential Therapeutic Strategies. Antioxidants (Basel) 2023; 12:antiox12051036. [PMID: 37237902 DOI: 10.3390/antiox12051036] [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: 03/23/2023] [Revised: 04/20/2023] [Accepted: 04/29/2023] [Indexed: 05/28/2023] Open
Abstract
Peripheral facial nerve injury leads to changes in the expression of various neuroactive substances that affect nerve cell damage, survival, growth, and regeneration. In the case of peripheral facial nerve damage, the injury directly affects the peripheral nerves and induces changes in the central nervous system (CNS) through various factors, but the substances involved in these changes in the CNS are not well understood. The objective of this review is to investigate the biomolecules involved in peripheral facial nerve damage so as to gain insight into the mechanisms and limitations of targeting the CNS after such damage and identify potential facial nerve treatment strategies. To this end, we searched PubMed using keywords and exclusion criteria and selected 29 eligible experimental studies. Our analysis summarizes basic experimental studies on changes in the CNS following peripheral facial nerve damage, focusing on biomolecules that increase or decrease in the CNS and/or those involved in the damage, and reviews various approaches for treating facial nerve injury. By establishing the biomolecules in the CNS that change after peripheral nerve damage, we can expect to identify factors that play an important role in functional recovery from facial nerve damage. Accordingly, this review could represent a significant step toward developing treatment strategies for peripheral facial palsy.
Collapse
Affiliation(s)
- Jae-Min Lee
- Department of Otorhinolaryngology, Head & Neck Surgery, College of Medicine, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea
| | - You Jung Choi
- Department of Otorhinolaryngology, Head & Neck Surgery, College of Medicine, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea
| | - Myung Chul Yoo
- Department of Physical Medicine & Rehabilitation, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung Geun Yeo
- Department of Otorhinolaryngology, Head & Neck Surgery, College of Medicine, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea
| |
Collapse
|
22
|
Xu T, Shi Y, Zheng G, Zhang G. Diagnostic Potential of Two Novel Biomarkers for Neuromyelitis Optica Spectrum Disorder and Multiple Sclerosis. Diagnostics (Basel) 2023; 13:diagnostics13091572. [PMID: 37174963 PMCID: PMC10178292 DOI: 10.3390/diagnostics13091572] [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: 03/07/2023] [Revised: 04/07/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Currently, no tests can definitively diagnose and distinguish neuromyelitis optica spectrum disorder (NMOSD) from multiple sclerosis (MS). METHODS Initially, cerebrospinal fluid (CSF) proteomics were employed to uncover the novel biomarkers that differentiate NMOSD from MS into cohorts of 10 MS and 10 NMOSD patients. Subsequently, screening biomarkers were validated using an enzyme-linked immunosorbent assay method and CSF and serum samples from 20 MS patients, 20 NMOSD patients, 20 non-inflammatory neurological controls, and 20 healthy controls. RESULTS In study cohort, insulin-like growth factor-binding protein 7 (IGFBP7) and lysosome-associated membrane glycoprotein 2 (LAMP2) were screened. In validation cohort, serum and CSF IGFBP7 not only exhibited higher levels in MS and NMOSD patients than controls, but also had greatest area under the curve (AUC, above or equal to 0.8) in MS and NMOSD diagnoses. Serum IGFBP7 (0.945) and CSF IGFBP7 (0.890) also had the greatest AUCs for predicting MS progression, while serum LAMP2 had a moderate curve (0.720). CONCLUSIONS IGFBP7 was superior in diagnosing MS and NMOSD, and IGFBP7 and serum LAMP2 performed exceptionally well in predicting the MS progression. These results offered reasons for further investigations into the functions of IGFBP7 and LAMP2 in MS and NMOSD.
Collapse
Affiliation(s)
- Ting Xu
- Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- NMPA Key Laboratory for Quality Control of In Vitro Diagnostics, Beijing 100070, China
- Beijing Engineering Research Center of Immunological Reagents Clinical Research, Beijing 100070, China
| | - Yijun Shi
- Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- NMPA Key Laboratory for Quality Control of In Vitro Diagnostics, Beijing 100070, China
- Beijing Engineering Research Center of Immunological Reagents Clinical Research, Beijing 100070, China
| | - Guanghui Zheng
- Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- NMPA Key Laboratory for Quality Control of In Vitro Diagnostics, Beijing 100070, China
- Beijing Engineering Research Center of Immunological Reagents Clinical Research, Beijing 100070, China
| | - Guojun Zhang
- Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- NMPA Key Laboratory for Quality Control of In Vitro Diagnostics, Beijing 100070, China
- Beijing Engineering Research Center of Immunological Reagents Clinical Research, Beijing 100070, China
| |
Collapse
|
23
|
Kitamura K, Homma T, Sohel MSH, Fuyuki A, Miyawaki S, Onouchi S, Saito S. Expression patterns of prosaposin and its receptors, G protein-coupled receptor (GPR) 37 and GPR37L1, in the mouse olfactory organ. Tissue Cell 2023; 82:102093. [PMID: 37075680 DOI: 10.1016/j.tice.2023.102093] [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: 01/29/2023] [Revised: 03/24/2023] [Accepted: 04/14/2023] [Indexed: 04/21/2023]
Abstract
Prosaposin is a glycoprotein conserved widely in vertebrates, because it is a precursor for saposins that are required for normal lysosomal function and thus for autophagy, and acts as a neurotrophic factor. Most tetrapods possess two kinds of olfactory neuroepithelia, namely, the olfactory epithelium (OE) and the vomeronasal epithelium (VNE). This study examined the expression patterns of prosaposin and its candidate receptors, G protein-coupled receptor (GPR) 37 and GPR37L1, in mouse OE and VNE by immunofluorescence and in situ hybridization. Prosaposin immunoreactivity was observed in the olfactory receptor neurons, vomeronasal receptor neurons, Bowman's gland (BG), and Jacobson's gland (JG). Prosaposin expression was mainly observed in mature neurons. Prosaposin mRNA expression was observed not only in these cells but also in the apical region of the VNE. GPR37 and GPR37L1 immunoreactivities were found only in the BG and/or the JG. Prosaposin was suggested to secrete and facilitate the autophagic activities of the neurons and modulate the mucus secretion in mouse olfactory organ.
Collapse
Affiliation(s)
- Kai Kitamura
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Takeshi Homma
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, Japan
| | - Md Shahriar Hasan Sohel
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, Japan
| | - Aimi Fuyuki
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Shingo Miyawaki
- Laboratory of Veterinary Surgery, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Sawa Onouchi
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan; Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, Japan
| | - Shouichiro Saito
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan; Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, Japan.
| |
Collapse
|
24
|
Tong J, Guan X, Jiang S, Sun L. A saposin domain-containing protein of tongue sole Cynoglossus semilaevis: Antimicrobial activity and mechanism. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 141:104633. [PMID: 36610645 DOI: 10.1016/j.dci.2023.104633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 06/17/2023]
Abstract
Prosaposin is a precursor that can be processed into four different saposins, designated as A, B, C, and D, which have multiple functions in mammals, including neuroprotection and immune modulation. The immune function of saposin in teleost remains largely unknown. In the present study, a saposin (SAP) domain-containing protein was identified in half-smooth tongue sole Cynoglossus semilaevis and named CsSDP. CsSDP harbors one SAP A domain and two SAP B domains. When expressed in HEK293T cells, CsSDP was specifically localized in the lysosome. When overexpressed in Escherichia coli, CsSDP markedly inhibited bacterial growth, and the inhibitory effect depended on two specific regions in the SAP A and SAP B domains. Two polypeptides (P32 and P30) derived from the above SAP A and B domains could bind to and inhibit the growth of both Gram-negative and Gram-positive bacteria. The ultrastructural analysis revealed that P32 and P30 killed target bacteria by disrupting the bacterial cell wall and inducing substantial release of cytoplasmic contents. These results shed new lights on the immune function of saposin domain-containing protein in teleost.
Collapse
Affiliation(s)
- Jiazhou Tong
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China; CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Xiaolu Guan
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
| | - Shuai Jiang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China.
| | - Li Sun
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China.
| |
Collapse
|
25
|
Pandey MK. Exploring Pro-Inflammatory Immunological Mediators: Unraveling the Mechanisms of Neuroinflammation in Lysosomal Storage Diseases. Biomedicines 2023; 11:biomedicines11041067. [PMID: 37189685 DOI: 10.3390/biomedicines11041067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 04/05/2023] Open
Abstract
Lysosomal storage diseases are a group of rare and ultra-rare genetic disorders caused by defects in specific genes that result in the accumulation of toxic substances in the lysosome. This excess accumulation of such cellular materials stimulates the activation of immune and neurological cells, leading to neuroinflammation and neurodegeneration in the central and peripheral nervous systems. Examples of lysosomal storage diseases include Gaucher, Fabry, Tay–Sachs, Sandhoff, and Wolman diseases. These diseases are characterized by the accumulation of various substrates, such as glucosylceramide, globotriaosylceramide, ganglioside GM2, sphingomyelin, ceramide, and triglycerides, in the affected cells. The resulting pro-inflammatory environment leads to the generation of pro-inflammatory cytokines, chemokines, growth factors, and several components of complement cascades, which contribute to the progressive neurodegeneration seen in these diseases. In this study, we provide an overview of the genetic defects associated with lysosomal storage diseases and their impact on the induction of neuro-immune inflammation. By understanding the underlying mechanisms behind these diseases, we aim to provide new insights into potential biomarkers and therapeutic targets for monitoring and managing the severity of these diseases. In conclusion, lysosomal storage diseases present a complex challenge for patients and clinicians, but this study offers a comprehensive overview of the impact of these diseases on the central and peripheral nervous systems and provides a foundation for further research into potential treatments.
Collapse
Affiliation(s)
- Manoj Kumar Pandey
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, Cincinnati, OH 45229-3026, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0515, USA
| |
Collapse
|
26
|
Fuyuki A, Yamamoto S, Sohel MSH, Homma T, Kitamura K, Onouchi S, Saito S. Expression of prosaposin and its G protein-coupled receptor (GPR) 37 in mouse cochlear and vestibular nuclei. J Vet Med Sci 2023; 85:266-270. [PMID: 36696997 PMCID: PMC10076190 DOI: 10.1292/jvms.22-0552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Prosaposin is a precursor of lysosomal hydrolases activator proteins, saposins, and also acts as a secretory protein that is not processed into saposins. Prosaposin elicits neurotrophic function via G protein-coupled receptor (GPR) 37, and prosaposin deficiency causes abnormal vestibuloauditory end-organ development. In this study, immunohistochemistry was used to examine prosaposin and GPR37 expression patterns in the mouse cochlear and vestibular nuclei. Prosaposin immunoreactivity was observed in neurons and glial cells in both nuclei. GPR37 immunoreactivity was observed in only some neurons, and its immunoreactivity in the vestibular nucleus was weaker than that in the cochlear nucleus. This study suggests a possibility that prosaposin deficiency affects not only the end-organs but also the first center of the vestibuloauditory system.
Collapse
Affiliation(s)
- Aimi Fuyuki
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Saki Yamamoto
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Md Shahriar Hasan Sohel
- Laboratory of Veterinary Anatomy, The Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Takeshi Homma
- Laboratory of Veterinary Anatomy, The Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Kai Kitamura
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Sawa Onouchi
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan.,Laboratory of Veterinary Anatomy, The Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Shouichiro Saito
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan.,Laboratory of Veterinary Anatomy, The Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| |
Collapse
|
27
|
Bhat V, Thergaonkar R, Thakur M, Rajkamal T. Combined saposin deficiency: A rare occurrence. Med J Armed Forces India 2023; 79:238-240. [PMID: 36969110 PMCID: PMC10037043 DOI: 10.1016/j.mjafi.2021.01.024] [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: 05/04/2020] [Accepted: 01/23/2021] [Indexed: 01/20/2023] Open
Abstract
Combined saposin deficiency (OMIM #611721), an exceedingly rare lysosomal storage disorder, is caused by a mutation in the gene PSAP. This gene encodes a protein, prosaposin, that cleaves into four constituent proteins, each of which has a role as a cofactor for the enzymes whose deficiency results in Krabbe disease, metachromatic leukodystrophy, Gaucher disease, and Farber disease, respectively. Intact prosaposin itself is essential for neuronal survival. The typical manifestation of combined saposin deficiency is of severe neurological features in the neonatal period, hepatosplenomegaly, thrombocytopenia, and early death. We report, to the best of our knowledge, the first Indian case with these clinical manifestations and confirmation by genetic and enzymatic testing.
Collapse
Affiliation(s)
- Vivek Bhat
- Senior Advisor (Pediatrics) & Neonatologist, INHS Kalyani, Visakhapatnam, India
| | - R.W. Thergaonkar
- Senior Advisor (Pediatrics) & Pediatric Nephrologist, INHS Asvini, Mumbai, India
| | - Manisha Thakur
- Ex-Classified Specialist (Pediatrics), INHS Kalyani, Visakhapatnam, India
| | - T. Rajkamal
- Classified Specialist (Dermatology), INHS Kalyani, Visakhapatnam, India
| |
Collapse
|
28
|
Feng R, Muraleedharan Saraswathy V, Mokalled MH, Cavalli V. Self-renewing macrophages in dorsal root ganglia contribute to promote nerve regeneration. Proc Natl Acad Sci U S A 2023; 120:e2215906120. [PMID: 36763532 PMCID: PMC9963351 DOI: 10.1073/pnas.2215906120] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 01/07/2023] [Indexed: 02/11/2023] Open
Abstract
Sensory neurons located in dorsal root ganglia (DRG) convey sensory information from peripheral tissue to the brain. After peripheral nerve injury, sensory neurons switch to a regenerative state to enable axon regeneration and functional recovery. This process is not cell autonomous and requires glial and immune cells. Macrophages in the DRG (DRGMacs) accumulate in response to nerve injury, but their origin and function remain unclear. Here, we mapped the fate and response of DRGMacs to nerve injury using macrophage depletion, fate-mapping, and single-cell transcriptomics. We identified three subtypes of DRGMacs after nerve injury in addition to a small population of circulating bone-marrow-derived precursors. Self-renewing macrophages, which proliferate from local resident macrophages, represent the largest population of DRGMacs. The other two subtypes include microglia-like cells and macrophage-like satellite glial cells (SGCs) (Imoonglia). We show that self-renewing DRGMacs contribute to promote axon regeneration. Using single-cell transcriptomics data and CellChat to simulate intercellular communication, we reveal that macrophages express the neuroprotective and glioprotective ligand prosaposin and communicate with SGCs via the prosaposin receptor GPR37L1. These data highlight that DRGMacs have the capacity to self-renew, similarly to microglia in the Central nervous system (CNS) and contribute to promote axon regeneration. These data also reveal the heterogeneity of DRGMacs and their potential neuro- and glioprotective roles, which may inform future therapeutic approaches to treat nerve injury.
Collapse
Affiliation(s)
- Rui Feng
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO63110
| | - Vishnu Muraleedharan Saraswathy
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO63110
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - Mayssa H. Mokalled
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO63110
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO63110
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO63110
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO63110
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO63110
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO63110
| |
Collapse
|
29
|
Chu Y, Jia S, Xu K, Liu Q, Mai L, Liu J, Fan W, Huang F. Single-cell transcriptomic profile of satellite glial cells in trigeminal ganglion. Front Mol Neurosci 2023; 16:1117065. [PMID: 36818656 PMCID: PMC9932514 DOI: 10.3389/fnmol.2023.1117065] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Satellite glial cells (SGCs) play an important role in regulating the function of trigeminal ganglion (TG) neurons. Multiple mediators are involved in the bidirectional communication between SGCs and neurons in different physiological and pathological states. However, molecular insights into the transcript characteristics of SGCs are limited. Moreover, little is known about the heterogeneity of SGCs in TG, and a more in-depth understanding of the interactions between SGCs and neuron subtypes is needed. Here we show the single-cell RNA sequencing (scRNA-seq) profile of SGCs in TG under physiological conditions. Our results demonstrate TG includes nine types of cell clusters, such as neurons, SGCs, myeloid Schwann cells (mSCs), non-myeloid Schwann cells (nmSCs), immune cells, etc., and the corresponding markers are also presented. We reveal the signature gene expression of SGCs, mSCs and nmSCs in the TG, and analyze the ligand-receptor pairs between neuron subtypes and SGCs in the TG. In the heterogeneity analysis of SGCs, four SGCs subtypes are identified, including subtypes enriched for genes associated with extracellular matrix organization, immediate early genes, interferon beta, and cell adhesion molecules, respectively. Our data suggest the molecular characteristics, heterogeneity of SGCs, and bidirectional interactions between SGCs and neurons, providing a valuable resource for studying SGCs in the TG.
Collapse
Affiliation(s)
- Yanhao Chu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Shilin Jia
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Ke Xu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Qing Liu
- Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Lijia Mai
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jiawei Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wenguo Fan
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China,*Correspondence: Wenguo Fan, ; Fang Huang,
| | - Fang Huang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China,*Correspondence: Wenguo Fan, ; Fang Huang,
| |
Collapse
|
30
|
Cang Z, Zhao Y, Almet AA, Stabell A, Ramos R, Plikus MV, Atwood SX, Nie Q. Screening cell-cell communication in spatial transcriptomics via collective optimal transport. Nat Methods 2023; 20:218-228. [PMID: 36690742 PMCID: PMC9911355 DOI: 10.1038/s41592-022-01728-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/21/2022] [Indexed: 01/24/2023]
Abstract
Spatial transcriptomic technologies and spatially annotated single-cell RNA sequencing datasets provide unprecedented opportunities to dissect cell-cell communication (CCC). However, incorporation of the spatial information and complex biochemical processes required in the reconstruction of CCC remains a major challenge. Here, we present COMMOT (COMMunication analysis by Optimal Transport) to infer CCC in spatial transcriptomics, which accounts for the competition between different ligand and receptor species as well as spatial distances between cells. A collective optimal transport method is developed to handle complex molecular interactions and spatial constraints. Furthermore, we introduce downstream analysis tools to infer spatial signaling directionality and genes regulated by signaling using machine learning models. We apply COMMOT to simulation data and eight spatial datasets acquired with five different technologies to show its effectiveness and robustness in identifying spatial CCC in data with varying spatial resolutions and gene coverages. Finally, COMMOT identifies new CCCs during skin morphogenesis in a case study of human epidermal development.
Collapse
Affiliation(s)
- Zixuan Cang
- Department of Mathematics and Center for Research in Scientific Computation, North Carolina State University, Raleigh, NC, USA
| | - Yanxiang Zhao
- Department of Mathematics, The George Washington University, Washington, DC, USA
| | - Axel A Almet
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - Adam Stabell
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Raul Ramos
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Maksim V Plikus
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Scott X Atwood
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA.
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
31
|
Armando I, Cuevas S, Fan C, Kumar M, Izzi Z, Jose PA, Konkalmatt PR. G Protein-Coupled Receptor 37L1 Modulates Epigenetic Changes in Human Renal Proximal Tubule Cells. Int J Mol Sci 2022; 23:ijms232214456. [PMID: 36430934 PMCID: PMC9698582 DOI: 10.3390/ijms232214456] [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: 05/10/2022] [Revised: 10/19/2022] [Accepted: 10/31/2022] [Indexed: 11/23/2022] Open
Abstract
Renal luminal sodium transport is essential for physiological blood pressure control, and abnormalities in this process are strongly implicated in the pathogenesis of essential hypertension. Renal G protein-coupled receptors (GPCRs) are critical for the regulation of the reabsorption of essential nutrients, ions, and water from the glomerular filtrate. Recently, we showed that GPCR 37L1 (GPR37L1) is expressed on the apical membrane of renal proximal tubules (RPT) and regulates luminal sodium transport and blood pressure by modulating the function of the sodium proton exchanger 3 (NHE3). However, little is known about GPR37L1 intracellular signaling. Here, we show that GPR37L1 is localized to the nuclear membrane, in addition to the plasma membrane in human RPT cells. Furthermore, GPR37L1 signals via the PI3K/AKT/mTOR pathway to decrease the expression of DNA (cytosine-5)-methyltransferase 1 (DNMT1) and enhance NHE3 transcription. Overall, we demonstrate the direct role of a nuclear membrane GPCR in the regulation of renal sodium through epigenetic gene regulation.
Collapse
|
32
|
Decreased Prosaposin and Progranulin in the Cingulate Cortex Are Associated with Schizophrenia Pathophysiology. Int J Mol Sci 2022; 23:ijms231912056. [PMID: 36233357 PMCID: PMC9570388 DOI: 10.3390/ijms231912056] [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: 08/29/2022] [Revised: 10/02/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022] Open
Abstract
Prosaposin (PSAP) and progranulin (PGRN) are two lysosomal proteins that interact and modulate the metabolism of lipids, particularly sphingolipids. Alterations in sphingolipid metabolism have been found in schizophrenia. Genetic associations of PSAP and PGRN with schizophrenia have been reported. To further clarify the role of PSAP and PGRN in schizophrenia, we examined PSAP and PGRN levels in postmortem cingulate cortex tissue from healthy controls along with patients who had suffered from schizophrenia, bipolar disorder, or major depressive disorder. We found that PSAP and PGRN levels are reduced specifically in schizophrenia patients. To understand the role of PSAP in the cingulate cortex, we used an AAV strategy to knock down PSAP in neurons located in this region. Neuronal PSAP knockdown led to the downregulation of neuronal PGRN levels and behavioral abnormalities. Cingulate-PSAP-deficient mice exhibited increased anxiety-like behavior and impaired prepulse inhibition, as well as intact locomotion, working memory, and a depression-like state. The behavioral changes were accompanied by increased early growth response protein 1 (EGR-1) and activity-dependent cytoskeleton-associated protein (ARC) levels in the sensorimotor cortex and hippocampus, regions implicated in circuitry dysfunction in schizophrenia. In conclusion, PSAP and PGRN downregulation in the cingulate cortex is associated with schizophrenia pathophysiology.
Collapse
|
33
|
Jaago M, Rähni A, Pupina N, Pihlak A, Sadam H, Tuvikene J, Avarlaid A, Planken A, Planken M, Haring L, Vasar E, Baćević M, Lambert F, Kalso E, Pussinen P, Tienari PJ, Vaheri A, Lindholm D, Timmusk T, Ghaemmaghami AM, Palm K. Differential patterns of cross-reactive antibody response against SARS-CoV-2 spike protein detected for chronically ill and healthy COVID-19 naïve individuals. Sci Rep 2022; 12:16817. [PMID: 36207326 PMCID: PMC9540097 DOI: 10.1038/s41598-022-20849-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
Immunity to previously encountered viruses can alter response to unrelated pathogens. We reasoned that similar mechanism may also involve SARS-CoV-2 and thereby affect the specificity and the quality of the immune response against the virus. Here, we employed high-throughput next generation phage display method to explore the link between antibody immune response to previously encountered antigens and spike (S) glycoprotein. By profiling the antibody response in COVID-19 naïve individuals with a diverse clinical history (including cardiovascular, neurological, or oncological diseases), we identified 15 highly antigenic epitopes on spike protein that showed cross-reactivity with antigens of seasonal, persistent, latent or chronic infections from common human viruses. We observed varying degrees of cross-reactivity of different viral antigens with S in an epitope-specific manner. The data show that pre-existing SARS-CoV-2 S1 and S2 cross-reactive serum antibody is readily detectable in pre-pandemic cohort. In the severe COVID-19 cases, we found differential antibody response to the 15 defined antigenic and cross-reactive epitopes on spike. We also noted that despite the high mutation rates of Omicron (B.1.1.529) variants of SARS-CoV-2, some of the epitopes overlapped with the described mutations. Finally, we propose that the resolved epitopes on spike if targeted by re-called antibody response from SARS-CoV-2 infections or vaccinations can function in chronically ill COVID-19 naïve/unvaccinated individuals as immunogenic targets to boost antibodies augmenting the chronic conditions. Understanding the relationships between prior antigen exposure at the antibody epitope level and the immune response to subsequent infections with viruses from a different strain is paramount to guiding strategies to exit the COVID-19 pandemic.
Collapse
Affiliation(s)
- Mariliis Jaago
- Protobios LLC, Tallinn, Estonia
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Annika Rähni
- Protobios LLC, Tallinn, Estonia
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | | | | | - Helle Sadam
- Protobios LLC, Tallinn, Estonia
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Jürgen Tuvikene
- Protobios LLC, Tallinn, Estonia
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- DXLabs LLC, Tallinn, Estonia
| | - Annela Avarlaid
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Anu Planken
- North Estonia Medical Centre Foundation, Tallinn, Estonia
| | - Margus Planken
- North Estonia Medical Centre Foundation, Tallinn, Estonia
| | - Liina Haring
- Institute of Clinical Medicine, Psychiatry Clinic of Tartu University Hospital, University of Tartu, Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
- Center of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Miljana Baćević
- Dental Biomaterial Research Unit (d-BRU), Faculty of Medicine, University of Liege, Liege, Belgium
| | - France Lambert
- Department of Periodontology and Oral Surgery, Faculty of Medicine, University of Liege, Liege, Belgium
| | - Eija Kalso
- Department of Anaesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital, Helsinki, Finland
- SleepWell Research Programme, Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Pirkko Pussinen
- Oral and Maxillofacial Diseases, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Pentti J Tienari
- Translational Immunology Research Program, Department of Neurology, Neurocenter, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Antti Vaheri
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Dan Lindholm
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Tõnis Timmusk
- Protobios LLC, Tallinn, Estonia
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Amir M Ghaemmaghami
- Immunology and Immuno-Bioengineering Group, School of Life Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | | |
Collapse
|
34
|
Yan L, Chen S, Hou C, Lin J, Xiong W, Shen Y, Zhou T. Multi-omics analysis unravels dysregulated lysosomal function and lipid metabolism involved in sub-chronic particulate matter-induced pulmonary injury. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 836:155642. [PMID: 35525343 DOI: 10.1016/j.scitotenv.2022.155642] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/27/2022] [Accepted: 04/28/2022] [Indexed: 06/14/2023]
Abstract
Particulate matter (PM) is a huge environmental threat and is of major public concern. Oxidative stress and systemic inflammation are known factors that contribute to PM- related damage; however, a systematic understanding of the deleterious pulmonary effects of PM using multi-omics analysis is lacking. In this study, we performed transcriptomic, proteomic, and metabolomic analyses in a mouse model exposed to PM for three months to identify molecular changes in lung tissues. We identified 1690 genes, 326 proteins, and 67 metabolites exhibiting significant differences between PM-challenged and control mice (p < 0.05). Differentially expressed genes and proteins regulated in PM-challenged mice were involved in lipid metabolism and in the immune and inflammatory response processes. Moreover, a comprehensive analysis of transcript, protein, and metabolite datasets revealed that the genes, proteins, and metabolites in the PM-treated group were involved in lysosomal function and lipid metabolism. Specifically, Cathepsin D (Ctsd), Ferritin light chain (Ftl), Lactotransferrin (Ltf), Lipocalin 2 (Lcn2), and Prosaposin (Psap) were major proteins/genes associated with PM-induced pulmonary damage, while two lipid molecules PC (18:1(11Z)/16:0) and PA (16:0/18:1(11Z)) were major metabolites related to PM-induced pulmonary injury. In summary, lipid metabolism might be used as successful precautions and therapeutic targets in PM-induced pulmonary injury to maintain the stability of cellular lysosomal function.
Collapse
Affiliation(s)
- Lifeng Yan
- Department of Respiratory and Critical Care, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shangheng Chen
- Department of Forensic Medicine, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Chenchen Hou
- Department of Respiratory and Critical Care, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Junyi Lin
- Department of Forensic Medicine, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Weining Xiong
- Department of Respiratory and Critical Care, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China; Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yiwen Shen
- Department of Forensic Medicine, Shanghai Medical College of Fudan University, Shanghai 200032, China.
| | - Tianyu Zhou
- Department of Respiratory and Critical Care, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China; Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
35
|
Gomez-Arboledas A, Carvalho K, Balderrama-Gutierrez G, Chu SH, Liang HY, Schartz ND, Selvan P, Petrisko TJ, Pan MA, Mortazavi A, Tenner AJ. C5aR1 antagonism alters microglial polarization and mitigates disease progression in a mouse model of Alzheimer's disease. Acta Neuropathol Commun 2022; 10:116. [PMID: 35978440 PMCID: PMC9386996 DOI: 10.1186/s40478-022-01416-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/29/2022] [Indexed: 11/10/2022] Open
Abstract
Multiple studies have recognized the involvement of the complement cascade during Alzheimer’s disease pathogenesis. However, the specific role of C5a-C5aR1 signaling in the progression of this neurodegenerative disease is still not clear. Furthermore, its potential as a therapeutic target to treat AD still remains to be elucidated. Canonically, generation of the anaphylatoxin C5a as the result of complement activation and interaction with its receptor C5aR1 triggers a potent inflammatory response. Previously, genetic ablation of C5aR1 in a mouse model of Alzheimer’s disease exerted a protective effect by preventing cognitive deficits. Here, using PMX205, a potent, specific C5aR1 antagonist, in the Tg2576 mouse model of Alzheimer’s disease we show a striking reduction in dystrophic neurites in parallel with the reduced amyloid load, rescue of the excessive pre-synaptic loss associated with AD cognitive impairment and the polarization of microglial gene expression towards a DAM-like phenotype that are consistent with the neuroprotective effects seen. These data support the beneficial effect of a pharmacological inhibition of C5aR1 as a promising therapeutic approach to treat Alzheimer’s disease. Supportive of the safety of this treatment is the recent FDA-approval of another other C5a receptor 1 antagonist, Avacopan, as a treatment for autoimmune inflammatory diseases.
Collapse
Affiliation(s)
- Angela Gomez-Arboledas
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Klebea Carvalho
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | | | - Shu-Hui Chu
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Heidi Yahan Liang
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Nicole D Schartz
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Purnika Selvan
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Tiffany J Petrisko
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Miranda A Pan
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Andrea J Tenner
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA. .,Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA. .,Department of Pathology and Experimental Medicine, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
36
|
Gong L, Gu Y, Han X, Luan C, Liu C, Wang X, Sun Y, Zheng M, Fang M, Yang S, Xu L, Sun H, Yu B, Gu X, Zhou S. Spatiotemporal Dynamics of the Molecular Expression Pattern and Intercellular Interactions in the Glial Scar Response to Spinal Cord Injury. Neurosci Bull 2022; 39:213-244. [PMID: 35788904 PMCID: PMC9905408 DOI: 10.1007/s12264-022-00897-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/28/2022] [Indexed: 12/22/2022] Open
Abstract
Nerve regeneration in adult mammalian spinal cord is poor because of the lack of intrinsic regeneration of neurons and extrinsic factors - the glial scar is triggered by injury and inhibits or promotes regeneration. Recent technological advances in spatial transcriptomics (ST) provide a unique opportunity to decipher most genes systematically throughout scar formation, which remains poorly understood. Here, we first constructed the tissue-wide gene expression patterns of mouse spinal cords over the course of scar formation using ST after spinal cord injury from 32 samples. Locally, we profiled gene expression gradients from the leading edge to the core of the scar areas to further understand the scar microenvironment, such as neurotransmitter disorders, activation of the pro-inflammatory response, neurotoxic saturated lipids, angiogenesis, obstructed axon extension, and extracellular structure re-organization. In addition, we described 21 cell transcriptional states during scar formation and delineated the origins, functional diversity, and possible trajectories of subpopulations of fibroblasts, glia, and immune cells. Specifically, we found some regulators in special cell types, such as Thbs1 and Col1a2 in macrophages, CD36 and Postn in fibroblasts, Plxnb2 and Nxpe3 in microglia, Clu in astrocytes, and CD74 in oligodendrocytes. Furthermore, salvianolic acid B, a blood-brain barrier permeation and CD36 inhibitor, was administered after surgery and found to remedy fibrosis. Subsequently, we described the extent of the scar boundary and profiled the bidirectional ligand-receptor interactions at the neighboring cluster boundary, contributing to maintain scar architecture during gliosis and fibrosis, and found that GPR37L1_PSAP, and GPR37_PSAP were the most significant gene-pairs among microglia, fibroblasts, and astrocytes. Last, we quantified the fraction of scar-resident cells and proposed four possible phases of scar formation: macrophage infiltration, proliferation and differentiation of scar-resident cells, scar emergence, and scar stationary. Together, these profiles delineated the spatial heterogeneity of the scar, confirmed the previous concepts about scar architecture, provided some new clues for scar formation, and served as a valuable resource for the treatment of central nervous system injury.
Collapse
Affiliation(s)
- Leilei Gong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaoxiao Han
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Chengcheng Luan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Chang Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xinghui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yufeng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Mengru Zheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Mengya Fang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Shuhai Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Lai Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
37
|
Kaufmann M, Schaupp AL, Sun R, Coscia F, Dendrou CA, Cortes A, Kaur G, Evans HG, Mollbrink A, Navarro JF, Sonner JK, Mayer C, DeLuca GC, Lundeberg J, Matthews PM, Attfield KE, Friese MA, Mann M, Fugger L. Identification of early neurodegenerative pathways in progressive multiple sclerosis. Nat Neurosci 2022; 25:944-955. [PMID: 35726057 DOI: 10.1038/s41593-022-01097-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/16/2022] [Indexed: 11/08/2022]
Abstract
Progressive multiple sclerosis (MS) is characterized by unrelenting neurodegeneration, which causes cumulative disability and is refractory to current treatments. Drug development to prevent disease progression is an urgent clinical need yet is constrained by an incomplete understanding of its complex pathogenesis. Using spatial transcriptomics and proteomics on fresh-frozen human MS brain tissue, we identified multicellular mechanisms of progressive MS pathogenesis and traced their origin in relation to spatially distributed stages of neurodegeneration. By resolving ligand-receptor interactions in local microenvironments, we discovered defunct trophic and anti-inflammatory intercellular communications within areas of early neuronal decline. Proteins associated with neuronal damage in patient samples showed mechanistic concordance with published in vivo knockdown and central nervous system (CNS) disease models, supporting their causal role and value as potential therapeutic targets in progressive MS. Our findings provide a new framework for drug development strategies, rooted in an understanding of the complex cellular and signaling dynamics in human diseased tissue that facilitate this debilitating disease.
Collapse
Affiliation(s)
- Max Kaufmann
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Anna-Lena Schaupp
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Rosa Sun
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Department of Neurosurgery, Queen Elizabeth Hospital, Birmingham, UK
| | - Fabian Coscia
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Spatial Proteomics Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Calliope A Dendrou
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Adrian Cortes
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Gurman Kaur
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hayley G Evans
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Annelie Mollbrink
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| | - José Fernández Navarro
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| | - Jana K Sonner
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Christina Mayer
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, UK
| | - Joakim Lundeberg
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Kathrine E Attfield
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Mann
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Lars Fugger
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK.
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK.
| |
Collapse
|
38
|
Massimi M, Di Pietro C, La Sala G, Matteoni R. Mouse Mutants of Gpr37 and Gpr37l1 Receptor Genes: Disease Modeling Applications. Int J Mol Sci 2022; 23:ijms23084288. [PMID: 35457105 PMCID: PMC9025225 DOI: 10.3390/ijms23084288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 02/05/2023] Open
Abstract
The vertebrate G protein–coupled receptor 37 and G protein–coupled receptor 37-like 1 (GPR37 and GPR37L1) proteins have amino acid sequence homology to endothelin and bombesin-specific receptors. The prosaposin glycoprotein, its derived peptides, and analogues have been reported to interact with and activate both putative receptors. The GPR37 and GPR37L1 genes are highly expressed in human and rodent brains. GPR37 transcripts are most abundant in oligodendrocytes and in the neurons of the substantia nigra and hippocampus, while the GPR37L1 gene is markedly expressed in cerebellar Bergmann glia astrocytes. The human GPR37 protein is a substrate of parkin, and its insoluble form accumulates in brain samples from patients of inherited juvenile Parkinson’s disease. Several Gpr37 and Gpr37l1 mouse mutant strains have been produced and applied to extensive in vivo and ex vivo analyses of respective receptor functions and involvement in brain and other organ pathologies. The genotypic and phenotypic characteristics of the different mouse strains so far published are reported and discussed, and their current and proposed applications to human disease modeling are highlighted.
Collapse
|
39
|
Devireddy S, Ferguson SM. Efficient progranulin exit from the ER requires its interaction with prosaposin, a Surf4 cargo. J Cell Biol 2022; 221:e202104044. [PMID: 34919127 PMCID: PMC8689666 DOI: 10.1083/jcb.202104044] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 10/11/2021] [Accepted: 11/15/2021] [Indexed: 11/22/2022] Open
Abstract
Progranulin is a lysosomal protein whose haploinsufficiency causes frontotemporal dementia, while homozygous loss of progranulin causes neuronal ceroid lipofuscinosis, a lysosomal storage disease. The sensitivity of cells to progranulin deficiency raises important questions about how cells coordinate intracellular trafficking of progranulin to ensure its efficient delivery to lysosomes. In this study, we discover that progranulin interactions with prosaposin, another lysosomal protein, first occur within the lumen of the endoplasmic reticulum (ER) and are required for the efficient ER exit of progranulin. Mechanistically, we identify an interaction between prosaposin and Surf4, a receptor that promotes loading of lumenal cargos into COPII-coated vesicles, and establish that Surf4 is critical for the efficient export of progranulin and prosaposin from the ER. Collectively, this work demonstrates that a network of interactions occurring early in the secretory pathway promote the ER exit and subsequent lysosomal delivery of newly translated progranulin and prosaposin.
Collapse
Affiliation(s)
| | - Shawn M. Ferguson
- Departments of Cell Biology and Neuroscience, Program in Cellular Neuroscience, Neurodegeneration and Repair, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
40
|
An J, Zhang Y, Fudge AD, Lu H, Richardson WD, Li H. G protein-coupled receptor GPR37-like 1 regulates adult oligodendrocyte generation. Dev Neurobiol 2021; 81:975-984. [PMID: 34601807 DOI: 10.1002/dneu.22854] [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: 07/07/2021] [Revised: 09/12/2021] [Accepted: 09/29/2021] [Indexed: 02/01/2023]
Abstract
Oligodendrocytes (OLs) continue to be generated from OL precursors (OPs) in the adult mammalian brain. Adult-born OLs are believed to contribute to neural plasticity, learning and memory through a process of "adaptive myelination," but how adult OL generation and adaptive myelination are regulated remains unclear. Here, we report that the glia-specific G protein-coupled receptor 37-like 1 (GPR37L1) is expressed in subsets of OPs and newly formed immature OLs in adult mouse brain. We found that OP proliferation and differentiation are inhibited in the corpus callosum of adult Gpr37l1 knockout mice, leading to a reduction in the number of adult-born OLs. Our data raise the possibility that GPR37L1 is mechanistically involved in adult OL generation and adaptive myelination, and suggest that GPR37L1 might be a useful functional marker of OPs that are committed to OL differentiation.
Collapse
Affiliation(s)
- Jing An
- Faculty of Medical Sciences, Division of Medicine, Wolfson Institute for Biomedical Research, University College London, London, UK.,School of Basic Medical Sciences, Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yumeng Zhang
- Faculty of Medical Sciences, Division of Medicine, Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Alexander D Fudge
- Faculty of Medical Sciences, Division of Medicine, Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Haixia Lu
- School of Basic Medical Sciences, Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - William D Richardson
- Faculty of Medical Sciences, Division of Medicine, Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Huiliang Li
- Faculty of Medical Sciences, Division of Medicine, Wolfson Institute for Biomedical Research, University College London, London, UK
| |
Collapse
|
41
|
Mouat MA, Coleman JLJ, Wu J, Dos Remedios CG, Feneley MP, Graham RM, Smith NJ. Involvement of GPR37L1 in murine blood pressure regulation and human cardiac disease pathophysiology. Am J Physiol Heart Circ Physiol 2021; 321:H807-H817. [PMID: 34533400 DOI: 10.1152/ajpheart.00198.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 09/14/2021] [Indexed: 01/23/2023]
Abstract
Multiple mouse lines lacking the orphan G protein-coupled receptor, GPR37L1, have elicited disparate cardiovascular phenotypes. The first Gpr37l1 knockout mice study to be published reported a marked elevation in systolic blood pressure (SBP; ∼60 mmHg), revealing a potential therapeutic opportunity. The phenotype differed from our own independently generated knockout line, where male mice exhibited equivalent baseline blood pressure to wild type. Here, we attempted to reproduce the first study by characterizing the cardiovascular phenotype of both the original knockout and transgenic lines alongside a C57BL/6J control line, using the same method of blood pressure measurement. The present study supports the findings from our independently developed Gpr37l1 knockout line, finding that SBP and diastolic blood pressure (DBP) are not different in the original Gpr37l1 knockout male mice (SBP: 130.9 ± 5.3 mmHg; DBP: 90.7 ± 3.0 mmHg) compared with C57BL/6J mice (SBP: 123.1 ± 4.1 mmHg; DBP: 87.0 ± 2.7 mmHg). Instead, we attribute the apparent hypertension of the knockout line originally described to comparison with a seemingly hypotensive transgenic line (SBP 103.7 ± 5.0 mmHg; DBP 71.9 ± 3.7 mmHg). Additionally, we quantified myocardial GPR37L1 transcript in humans, which was suggested to be downregulated in cardiovascular disease. We found that GPR37L1 has very low native transcript levels in human myocardium and that expression is not different in tissue samples from patients with heart failure compared with sex-matched healthy control tissue. These findings indicate that cardiac GPR37L1 expression is unlikely to contribute to the pathophysiology of human heart failure.NEW & NOTEWORTHY This study characterizes systolic blood pressure (SBP) in a Gpr37l1 knockout mouse line, which was previously reported to have ∼60 mmHg higher SBP compared with a transgenic line. We observed only a ∼27 mmHg SBP difference between the lines. However, when compared with C57BL/6J mice, knockout mice showed no difference in SBP. We also investigated GPR37L1 mRNA abundance in human hearts and observed no difference between healthy and failing heart samples.
Collapse
Affiliation(s)
- Margaret A Mouat
- Molecular Pharmacology Laboratory, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- St Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - James L J Coleman
- Molecular Pharmacology Laboratory, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- St Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Jianxin Wu
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Cristobal G Dos Remedios
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Michael P Feneley
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Robert M Graham
- St Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Nicola J Smith
- Molecular Pharmacology Laboratory, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- St Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| |
Collapse
|
42
|
Yang L, Gong T, Shen H, Pei J, Zhang L, Zhang Q, Huang Y, Hu Z, Pan Z, Yang P, Lin L, Yu H. Precision N-Glycoproteomic Profiling of Murine Peritoneal Macrophages After Different Stimulations. Front Immunol 2021; 12:722293. [PMID: 34484231 PMCID: PMC8416091 DOI: 10.3389/fimmu.2021.722293] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
Macrophages are important immune cells that participate in both innate and adaptive immune responses, such as phagocytosis, recognition of molecular patterns, and activation of the immune response. In this study, murine peritoneal macrophages were isolated and then activated by LPS, HSV and VSV. Integrative proteomic and precision N-glycoproteomic profiling were conducted to assess the underlying macrophage activation. We identified a total of 587 glycoproteins, including 1239 glycopeptides, 526 monosaccharide components, and 8326 intact glycopeptides in glycoproteomics, as well as a total of 4496 proteins identified in proteomic analysis. These glycoproteins are widely involved in important biological processes, such as antigen presentation, cytokine production and glycosylation progression. Under the stimulation of the different pathogens, glycoproteins showed a dramatic change. We found that receptors in the Toll-like receptor pathway, such as Tlr2 and CD14, were increased under LPS and HSV stimulation. Glycosylation of those proteins was proven to influence their subcellular locations.
Collapse
Affiliation(s)
- Lujie Yang
- Institutes of Biomedical Sciences & Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Tianqi Gong
- Institutes of Biomedical Sciences & Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Huali Shen
- Institutes of Biomedical Sciences & Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Jiangnan Pei
- Obestetics & Gynecology Hospital, Fudan University, Shanghai, China
| | - Lei Zhang
- Institutes of Biomedical Sciences & Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Quanqing Zhang
- Department of Chemistry and Environmental Toxicology Graduate Program, University of California, Riverside, CA, United States
| | - Yuanyu Huang
- Institutes of Biomedical Sciences & Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Zuojian Hu
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ziyue Pan
- Institutes of Biomedical Sciences & Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Pengyuan Yang
- Institutes of Biomedical Sciences & Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Ling Lin
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, China
| | - Hongxiu Yu
- Institutes of Biomedical Sciences & Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| |
Collapse
|
43
|
Alonso-Gardón M, Elorza-Vidal X, Castellanos A, La Sala G, Armand-Ugon M, Gilbert A, Di Pietro C, Pla-Casillanis A, Ciruela F, Gasull X, Nunes V, Martínez A, Schulte U, Cohen-Salmon M, Marazziti D, Estévez R. Identification of the GlialCAM interactome: the G protein-coupled receptors GPRC5B and GPR37L1 modulate megalencephalic leukoencephalopathy proteins. Hum Mol Genet 2021; 30:1649-1665. [PMID: 34100078 PMCID: PMC8369841 DOI: 10.1093/hmg/ddab155] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/23/2022] Open
Abstract
Megalencephalic Leukoencephalopathy with subcortical Cysts (MLC) is a type of vacuolating leukodystrophy, which is mainly caused by mutations in MLC1 or GLIALCAM. The two MLC-causing genes encode for membrane proteins of yet unknown function that have been linked to the regulation of different chloride channels such as the ClC-2 and VRAC. To gain insight into the role of MLC proteins, we have determined the brain GlialCAM interacting proteome. The proteome includes different transporters and ion channels known to be involved in the regulation of brain homeostasis, proteins related to adhesion or signaling as several G protein-coupled receptors (GPCRs), including the orphan GPRC5B and the proposed prosaposin receptor GPR37L1. Focusing on these two GPCRs, we could validate that they interact directly with MLC proteins. The inactivation of Gpr37l1 in mice upregulated MLC proteins without altering their localization. Conversely, a reduction of GPRC5B levels in primary astrocytes downregulated MLC proteins, leading to an impaired activation of ClC-2 and VRAC. The interaction between the GPCRs and MLC1 was dynamically regulated upon changes in the osmolarity or potassium concentration. We propose that GlialCAM and MLC1 associate with different integral membrane proteins modulating their functions and acting as a recruitment site for various signaling components as the GPCRs identified here. We hypothesized that the GlialCAM/MLC1 complex is working as an adhesion molecule coupled to a tetraspanin-like molecule performing regulatory effects through direct binding or influencing signal transduction events.
Collapse
Affiliation(s)
- Marta Alonso-Gardón
- Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL - Institute of Neurosciences, Universitat de Barcelona, Barcelona 08036, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Xabier Elorza-Vidal
- Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL - Institute of Neurosciences, Universitat de Barcelona, Barcelona 08036, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Aida Castellanos
- Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL - Institute of Neurosciences, Universitat de Barcelona, Barcelona 08036, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Gina La Sala
- Institute of Biochemistry and Cell Biology, Italian National Research Council (CNR), Monterotondo Scalo, Rome I-00015, Italy
| | - Mercedes Armand-Ugon
- Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL - Institute of Neurosciences, Universitat de Barcelona, Barcelona 08036, Spain
| | - Alice Gilbert
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris F-75005, France
| | - Chiara Di Pietro
- Institute of Biochemistry and Cell Biology, Italian National Research Council (CNR), Monterotondo Scalo, Rome I-00015, Italy
| | - Adrià Pla-Casillanis
- Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL - Institute of Neurosciences, Universitat de Barcelona, Barcelona 08036, Spain
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona-IDIBELL, L'Hospitalet de Llobregat, Barcelona 08036, Spain
| | - Xavier Gasull
- Neurophysiology Laboratory, Department of Biomedicine, Medical School, Institute of Neurosciences, University of Barcelona-IDIBAPS, Casanova 143 Barcelona 08036, Spain
| | - Virginia Nunes
- Unitat de Genètica, Departament de Ciències Fisiològiques, Universitat de Barcelona, Laboratori de Genètica Molecular, Genes Disease and Therapy Program IDIBELL, L'Hospitalet de Llobregat 08036, Spain
| | - Albert Martínez
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | | | - Martine Cohen-Salmon
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris F-75005, France
| | - Daniela Marazziti
- Institute of Biochemistry and Cell Biology, Italian National Research Council (CNR), Monterotondo Scalo, Rome I-00015, Italy
| | - Raúl Estévez
- Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL - Institute of Neurosciences, Universitat de Barcelona, Barcelona 08036, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
44
|
Mattila SO, Tuhkanen HE, Lackman JJ, Konzack A, Morató X, Argerich J, Saftig P, Ciruela F, Petäjä-Repo UE. GPR37 is processed in the N-terminal ectodomain by ADAM10 and furin. FASEB J 2021; 35:e21654. [PMID: 34042202 DOI: 10.1096/fj.202002385rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 04/12/2021] [Accepted: 04/26/2021] [Indexed: 11/11/2022]
Abstract
GPR37 is an orphan G protein-coupled receptor (GPCR) implicated in several neurological diseases and important physiological pathways in the brain. We previously reported that its long N-terminal ectodomain undergoes constitutive metalloprotease-mediated cleavage and shedding, which have been rarely described for class A GPCRs. Here, we demonstrate that the protease that cleaves GPR37 at Glu167↓Gln168 is a disintegrin and metalloprotease 10 (ADAM10). This was achieved by employing selective inhibition, RNAi-mediated downregulation, and genetic depletion of ADAM10 in cultured cells as well as in vitro cleavage of the purified receptor with recombinant ADAM10. In addition, the cleavage was restored in ADAM10 knockout cells by overexpression of the wild type but not the inactive mutant ADAM10. Finally, postnatal conditional depletion of ADAM10 in mouse neuronal cells was found to reduce cleavage of the endogenous receptor in the brain cortex and hippocampus, confirming the physiological relevance of ADAM10 as a GPR37 sheddase. Additionally, we discovered that the receptor is subject to another cleavage step in cultured cells. Using site-directed mutagenesis, the site (Arg54↓Asp55) was localized to a highly conserved region at the distal end of the ectodomain that contains a recognition site for the proprotein convertase furin. The cleavage by furin was confirmed by using furin-deficient human colon carcinoma LoVo cells and proprotein convertase inhibitors. GPR37 is thus the first multispanning membrane protein that has been validated as an ADAM10 substrate and the first GPCR that is processed by both furin and ADAM10. The unconventional N-terminal processing may represent an important regulatory element for GPR37.
Collapse
Affiliation(s)
- S Orvokki Mattila
- Medical Research Center Oulu, Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Hanna E Tuhkanen
- Medical Research Center Oulu, Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Jarkko J Lackman
- Medical Research Center Oulu, Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Anja Konzack
- Medical Research Center Oulu, Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Xavier Morató
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - Josep Argerich
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - Paul Saftig
- Institute of Biochemistry, Kiel University, Kiel, Germany
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Ulla E Petäjä-Repo
- Medical Research Center Oulu, Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| |
Collapse
|
45
|
Sharoar MG, Palko S, Ge Y, Saido TC, Yan R. Accumulation of saposin in dystrophic neurites is linked to impaired lysosomal functions in Alzheimer's disease brains. Mol Neurodegener 2021; 16:45. [PMID: 34215298 PMCID: PMC8254260 DOI: 10.1186/s13024-021-00464-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 05/29/2021] [Indexed: 02/07/2023] Open
Abstract
Neuritic plaques in Alzheimer's disease (AD) brains refer to β-amyloid (Aβ) plaques surrounded by dystrophic neurites (DNs), activated microglia and reactive astrocytes. Most recently, we showed that DNs form sequentially in three layers during plaque growth. Although lysosomal proteins such as LAMP1 are found in DNs, it is not clear how many and how early lysosomal proteins are involved in forming neuritic plaques. To answer this unmet question, we examined APP knock-in (APPNL-G-F), 5xFAD and APP/PS1ΔE9 mouse brains and found that the lysosomal activator proteins saposins (SAPs) and LAMP1 were accumulated to surround Aβ plaques at the earliest stage, namely the 1st layer of DNs. Noticeably, lysosomal hydrolases were not detectable in these early DNs, suggesting that DNs at this early stage likely enrich dysfunctional lysosomes. In old AD mouse brains and in the later stage of human AD brains, SAP-C+-DNs and LAMP1+-DNs were gradually reduced in concomitant with the growth of amyloid plaques. Remarkably, the observed LAMP1 immunoreactivity near plaques in aged AD mouse and human brains were actually associated with disease-associated microglia rather than neuronal sources, likely reflecting more severely impaired lysosomal functions in neurons. Western blot analyses showed increased levels of SAP-C in AD mouse brains, and Aβ oligomers induced elevated levels of SAP-C in cellular assays. The elevated protein levels of SAP-C in AD mouse brains during plaque growth potentially contributed lysosomal membrane leakage and loss of hydrolases. Together, our study indicates that lysosomal functions are impaired by being entrapped in DNs early during plaque growth, and this may viciously facilitate growth of amyloid plaques.
Collapse
Affiliation(s)
- Md Golam Sharoar
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Sarah Palko
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Yingying Ge
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako-shi, Saitama, Japan
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06032, USA.
| |
Collapse
|
46
|
Do J, Perez G, Berhe B, Tayebi N, Sidransky E. Behavioral Phenotyping in a Murine Model of GBA1-Associated Parkinson Disease. Int J Mol Sci 2021; 22:ijms22136826. [PMID: 34202076 PMCID: PMC8267726 DOI: 10.3390/ijms22136826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/11/2021] [Accepted: 06/22/2021] [Indexed: 01/15/2023] Open
Abstract
Mutations in GBA1, the gene encoding glucocerebrosidase, are common genetic risk factors for Parkinson disease (PD). While the mechanism underlying this relationship is unclear, patients with GBA1-associated PD often have an earlier onset and faster progression than idiopathic PD. Previously, we modeled GBA1-associated PD by crossing gba haploinsufficient mice with mice overexpressing a human mutant α-synuclein transgene (SNCAA53T), observing an earlier demise, shorter life span and faster symptom progression, although behavioral testing was not performed. To assess whether gba+/−//SNCAA53T mice exhibit a prodromal behavioral phenotype, we studied three cardinal PD features: olfactory discrimination, memory dysfunction, and motor function. The longitudinal performance of gba+/−//SNCAA53T (n = 8), SNCAA53T (n = 9), gba+/− (n = 10) and wildtype (n = 6) mice was evaluated between ages 8 and 23 months using the buried pellet test, novel object recognition test and the beam walk. Fifteen-month-old gba+/−//SNCAA53T mice showed more olfactory and motor deficits than wildtype mice. However, differences between gba+/−//SNCAA53T and SNCAA53T mice generally did not reach statistical significance, possibly due to small sample sizes. Furthermore, while gba haploinsufficiency leads to a more rapid demise, this might not result in an earlier prodromal stage, and other factors, including aging, oxidative stress and epigenetics, may contribute to the more fulminant disease course.
Collapse
Affiliation(s)
| | | | | | - Nahid Tayebi
- Correspondence: (N.T.); (E.S.); Tel.: +1-301-496-0373 (N.T.); +1-301-451-0901 (E.S.)
| | - Ellen Sidransky
- Correspondence: (N.T.); (E.S.); Tel.: +1-301-496-0373 (N.T.); +1-301-451-0901 (E.S.)
| |
Collapse
|
47
|
Differential Regulation of Gonadotropins as Revealed by Transcriptomes of Distinct LH and FSH Cells of Fish Pituitary. Int J Mol Sci 2021; 22:ijms22126478. [PMID: 34204216 PMCID: PMC8234412 DOI: 10.3390/ijms22126478] [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: 05/11/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 02/08/2023] Open
Abstract
From mammals to fish, reproduction is driven by luteinizing hormone (LH) and follicle-stimulating hormone (FSH) temporally secreted from the pituitary gland. Teleost fish are an excellent model for addressing the unique regulation and function of each gonadotropin cell since, unlike mammals, they synthesize and secrete LH and FSH from distinct cells. Only very distant vertebrate classes (such as fish and birds) demonstrate the mono-hormonal strategy, suggesting a potential convergent evolution. Cell-specific transcriptome analysis of double-labeled transgenic tilapia expressing GFP and RFP in LH or FSH cells, respectively, yielded genes specifically enriched in each cell type, revealing differences in hormone regulation, receptor expression, cell signaling, and electrical properties. Each cell type expresses a unique GPCR signature that reveals the direct regulation of metabolic and homeostatic hormones. Comparing these novel transcriptomes to that of rat gonadotrophs revealed conserved genes that might specifically contribute to each gonadotropin activity in mammals, suggesting conserved mechanisms controlling the differential regulation of gonadotropins in vertebrates.
Collapse
|
48
|
Chen Z, Yu Q, Yu Q, Johnson J, Shipman R, Zhong X, Huang J, Asthana S, Carlsson C, Okonkwo O, Li L. In-depth Site-specific Analysis of N-glycoproteome in Human Cerebrospinal Fluid and Glycosylation Landscape Changes in Alzheimer's Disease. Mol Cell Proteomics 2021; 20:100081. [PMID: 33862227 PMCID: PMC8724636 DOI: 10.1016/j.mcpro.2021.100081] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/02/2021] [Accepted: 04/03/2021] [Indexed: 01/22/2023] Open
Abstract
As the body fluid that directly interchanges with the extracellular fluid of the central nervous system (CNS), cerebrospinal fluid (CSF) serves as a rich source for CNS-related disease biomarker discovery. Extensive proteome profiling has been conducted for CSF, but studies aimed at unraveling site-specific CSF N-glycoproteome are lacking. Initial efforts into site-specific N-glycoproteomics study in CSF yield limited coverage, hindering further experimental design of glycosylation-based disease biomarker discovery in CSF. In the present study, we have developed an N-glycoproteomic approach that combines enhanced N-glycopeptide sequential enrichment by hydrophilic interaction chromatography (HILIC) and boronic acid enrichment with electron transfer and higher-energy collision dissociation (EThcD) for large-scale intact N-glycopeptide analysis. The application of the developed approach to the analyses of human CSF samples enabled identifications of a total of 2893 intact N-glycopeptides from 511 N-glycosites and 285 N-glycoproteins. To our knowledge, this is the largest site-specific N-glycoproteome dataset reported for CSF to date. Such dataset provides molecular basis for a better understanding of the structure-function relationships of glycoproteins and their roles in CNS-related physiological and pathological processes. As accumulating evidence suggests that defects in glycosylation are involved in Alzheimer's disease (AD) pathogenesis, in the present study, a comparative in-depth N-glycoproteomic analysis was conducted for CSF samples from healthy control and AD patients, which yielded a comparable N-glycoproteome coverage but a distinct expression pattern for different categories of glycoforms, such as decreased fucosylation in AD CSF samples. Altered glycosylation patterns were detected for a number of N-glycoproteins including alpha-1-antichymotrypsin, ephrin-A3 and carnosinase CN1 etc., which serve as potentially interesting targets for further glycosylation-based AD study and may eventually lead to molecular elucidation of the role of glycosylation in AD progression.
Collapse
Affiliation(s)
- Zhengwei Chen
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, USA
| | - Qinying Yu
- School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
| | - Qing Yu
- School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
| | - Jillian Johnson
- School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
| | - Richard Shipman
- Department of Applied Science, University of Wisconsin-Stout, Menomonie, Wisconsin, USA
| | - Xiaofang Zhong
- School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
| | - Junfeng Huang
- School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
| | - Sanjay Asthana
- School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Cynthia Carlsson
- School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Ozioma Okonkwo
- School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin, USA; School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA.
| |
Collapse
|
49
|
Aslam M, Kandasamy N, Ullah A, Paramasivam N, Öztürk MA, Naureen S, Arshad A, Badshah M, Khan K, Wajid M, Abbasi R, Ilyas M, Eils R, Schlesner M, Wade RC, Ahmad N, von Engelhardt J. Putative second hit rare genetic variants in families with seemingly GBA-associated Parkinson's disease. NPJ Genom Med 2021; 6:2. [PMID: 33402667 PMCID: PMC7785741 DOI: 10.1038/s41525-020-00163-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 11/12/2020] [Indexed: 11/08/2022] Open
Abstract
Rare variants in the beta-glucocerebrosidase gene (GBA1) are common genetic risk factors for alpha synucleinopathy, which often manifests clinically as GBA-associated Parkinson's disease (GBA-PD). Clinically, GBA-PD closely mimics idiopathic PD, but it may present at a younger age and often aggregates in families. Most carriers of GBA variants are, however, asymptomatic. Moreover, symptomatic PD patients without GBA variant have been reported in families with seemingly GBA-PD. These observations obscure the link between GBA variants and PD pathogenesis and point towards a role for unidentified additional genetic and/or environmental risk factors or second hits in GBA-PD. In this study, we explored whether rare genetic variants may be additional risk factors for PD in two families segregating the PD-associated GBA1 variants c.115+1G>A (ClinVar ID: 93445) and p.L444P (ClinVar ID: 4288). Our analysis identified rare genetic variants of the HSP70 co-chaperone DnaJ homolog subfamily B member 6 (DNAJB6) and lysosomal protein prosaposin (PSAP) as additional factors possibly influencing PD risk in the two families. In comparison to the wild-type proteins, variant DNAJB6 and PSAP proteins show altered functions in the context of cellular alpha-synuclein homeostasis when expressed in reporter cells. Furthermore, the segregation pattern of the rare variants in the genes encoding DNAJB6 and PSAP indicated a possible association with PD in the respective families. The occurrence of second hits or additional PD cosegregating rare variants has important implications for genetic counseling in PD families with GBA1 variant carriers and for the selection of PD patients for GBA targeted treatments.
Collapse
Affiliation(s)
- Muhammad Aslam
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| | - Nirosiya Kandasamy
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Anwar Ullah
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan
- Department of Biochemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - Nagarajan Paramasivam
- Heidelberg Center for Personalized Oncology (DKFZ-HIPO), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mehmet Ali Öztürk
- Molecular and Cellular Modeling Group, Heidelberg Institute of Theoretical Studies (HITS), Heidelberg, Germany
- The Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104, Freiburg, Germany
| | - Saima Naureen
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Department of Zoology, PMAS-Arid Agriculture University, Rawalpindi, Pakistan
| | - Abida Arshad
- Department of Zoology, PMAS-Arid Agriculture University, Rawalpindi, Pakistan
| | - Mazhar Badshah
- Department of Neurology, Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad, Pakistan
| | - Kafaitullah Khan
- Department of Microbiology, University of Balochistan, Quetta, Pakistan
| | - Muhammad Wajid
- Department of Biological Sciences, University of Okara, Okara, Pakistan
| | - Rashda Abbasi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan
| | - Muhammad Ilyas
- Faculty of Mechanical Engineering, GIK Institute of Engineering Sciences and Technology, Topi, 23460, Pakistan
| | - Roland Eils
- Center for Digital Health, Berlin Institute of Health and Charité Universitätsmedizin Berlin, Berlin, Germany
- Health Data Science Unit, Bioquant, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Matthias Schlesner
- Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rebecca C Wade
- Molecular and Cellular Modeling Group, Heidelberg Institute of Theoretical Studies (HITS), Heidelberg, Germany
- Center for Molecular Biology of the University of Heidelberg (ZMBH), DKFZ-ZMBH Alliance, and Interdisciplinary Center for Scientific Computing (IWR), Heidelberg, Germany
| | - Nafees Ahmad
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan
| | - Jakob von Engelhardt
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
50
|
Kaczynski P, Bauersachs S, Goryszewska E, Baryla M, Waclawik A. Synergistic action of estradiol and PGE2 on endometrial transcriptome in vivo resembles pregnancy effects better than estradiol alone†. Biol Reprod 2020; 104:818-834. [PMID: 33354726 DOI: 10.1093/biolre/ioaa230] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/10/2020] [Accepted: 12/16/2020] [Indexed: 01/01/2023] Open
Abstract
Successful pregnancy establishment in mammals depends on numerous interactions between embryos and the maternal organism. Estradiol-17β (E2) is the primary embryonic signal in the pig, and its importance has been questioned recently. However, E2 is not the only molecule of embryonic origin. In pigs, prostaglandin E2 (PGE2) is abundantly synthesized and secreted by conceptuses and endometrium. The present study aimed to determine the role of PGE2 and its simultaneous action with E2 in changes in porcine endometrial transcriptome during pregnancy establishment. The effects of PGE2 and PGE2 acting with E2 were studied using an in vivo model of intrauterine hormone infusions, and were compared to the effects of E2 alone and conceptuses' presence on day 12 of pregnancy. The endometrial transcriptome was profiled using gene expression microarrays followed by statistical analyses. Downstream analyses were performed using bioinformatics tools. Differential expression of selected genes was verified by quantitative polymerase chain reaction. Microarray analysis revealed 2413 differentially expressed genes (DEGs) in the endometrium treated simultaneously with PGE2 and E2 (P < 0.01). No significant effect of PGE2 administered alone on endometrial transcriptome was detected. Gene ontology annotations enriched for DEGs were related to multiple processes such as: focal adhesion, vascularization, cell migration and proliferation, glucose metabolism, tissue remodeling, and activation of immune response. Simultaneous administration of E2 and PGE2 induced more changes within endometrial transcriptome characteristic to pregnancy than infusion of E2 alone. The present findings suggest that synergistic action of estradiol-17β and PGE2 resembles the effects of pregnancy on endometrial transcriptome better than E2 alone.
Collapse
Affiliation(s)
- Piotr Kaczynski
- Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Stefan Bauersachs
- Vetsuisse Faculty, Institute of Veterinary Anatomy, Functional Genomics, University of Zurich, Zurich, Switzerland
| | - Ewelina Goryszewska
- Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Monika Baryla
- Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Agnieszka Waclawik
- Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|