1
|
Ge Y, Teng R, Jia Z, Li Y, Lu Y, Yuan J. Elevated NGF provokes decidual lipid peroxidation and promotes preterm birth in mice. J Transl Med 2025; 23:481. [PMID: 40296138 PMCID: PMC12036137 DOI: 10.1186/s12967-025-06424-3] [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: 02/12/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Preterm birth (PTB) is a major cause of neonatal morbidity and mortality worldwide, with infection and inflammation being the most common triggers. However, many cases of preterm labor have unknown causes. The maternal decidua is a highly dynamic and heterogeneous region, serving as the nexus at the maternal-fetal interface, connecting the mother and the fetus. Increasing evidence suggests that the maternal decidua plays a crucial role in the initiation of labor. Nerve growth factor (NGF), is an important member of the neurotrophin family and is identified to play a crucial role in initiating the decidual response. METHODS To investigate whether NGF contributes to preterm birth via lipid peroxidation-dependent pathways, we selected both NGF and erastin (a pharmacological lipid peroxidation inducer) for parallel experimental treatments to examine how these pathways mediate the initiation of parturition. Mice were administered intraperitoneal injections of NGF and erastin. Gestational durations less than 19.5 days were classified as preterm birth. This study employed biological technologies and experimental methods to explore the initiation of delivery and the associated signaling pathways. RESULTS Elevated NGF levels in late-stage pregnancy increased the incidence of preterm birth in mice, independent of decidual senescence, placenta abnormal structure and ovarian dysfunction. Instead, NGF treatment activated lipid peroxidation and upregulated inflammatory markers in maternal decidua, particularly cyclooxygenase enzymes, which are critical for labor initiation. Notably, administration of erastin corroborated these findings, leading to similar outcomes in preterm labor. CONCLUSIONS This study reveals the pivotal role of NGF signaling in promoting excessive lipid peroxidation to disrupt decidual homeostasis and ultimately triggering preterm labor in mice.
Collapse
Affiliation(s)
- Yuhang Ge
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Ruxin Teng
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Zhaoyu Jia
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yongyue Li
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yafang Lu
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jia Yuan
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
2
|
Bogacheva PO, Potapova DA, Gaydukov AE. Sortilin and L-type Calcium Channels May be Involved in the Unusual Mechanism of proBDNF Signaling in Regenerating Mouse Neuromuscular Junctions. Neurochem Res 2025; 50:104. [PMID: 39998597 DOI: 10.1007/s11064-025-04360-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/17/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
proBDNF and its main proteolytic product BDNF play crucial roles in maturation of neuromuscular junctions during development or reinnervation. We investigated the mechanisms of acute proBDNF effects on synaptic transmission in mouse motor synapses regenerating after nerve crush. The cleavage-resistant proBDNF mimicked the previously shown effect of cleavable proBDNF- GIRK-mediated decrease in the miniature endplate potential (MEPP) frequency accompanied by slight hyperpolarization of postsynaptic membrane. Remarkably, this effect did not utilize canonical proBDNF signaling pathway since inhibition of either p75 receptors with LM11A-31 or sortilin with AF38469 was not able to prevent it. Without sortilin activity, proBDNF downregulated the quantal content of multiquantal endplate potentials (EPP). This non-canonical action of proneurotrophin via TrkB receptors highlights the important role of sortilin as a safeguard preventing the spread of the negative effect of proBDNF on the evoked neurotransmitter release in regenerating motor synapses. In the absence of sortilin activity L-type calcium channels emerged as the key players providing proBDNF-induced decrease of EPP quantal content, while they were not involved in proBDNF-induced decrease of MEPP frequency. Sortilin-independent but TrkB- and GIRK-mediated inhibition of spontaneous release by proBDNF was not associated with the activity of acetylcholine (M2) or purinergic (A1 and P2Y13) metabotropic receptors. We propose that depending on sortilin involvement, proBDNF selectively affects spontaneous or evoked quantal neurotransmitter release via different branches of signaling pathway that ensure the presynaptic activation of GIRK or L-type calcium channels, respectively.
Collapse
Affiliation(s)
- P O Bogacheva
- Faculty of Biology, Department of Human and Animal Physiology, Lomonosov Moscow State University, Moscow, Russian Federation.
| | - D A Potapova
- Faculty of Biology, Department of Human and Animal Physiology, Lomonosov Moscow State University, Moscow, Russian Federation
| | - A E Gaydukov
- Faculty of Biology, Department of Human and Animal Physiology, Lomonosov Moscow State University, Moscow, Russian Federation
| |
Collapse
|
3
|
Staib-Lasarzik I, Gölz C, Bobkiewiecz W, Somnuke P, Sebastiani A, Thal SC, Schäfer MK. Sortilin is dispensable for secondary injury processes following traumatic brain injury in mice. Heliyon 2024; 10:e35198. [PMID: 39170542 PMCID: PMC11336488 DOI: 10.1016/j.heliyon.2024.e35198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024] Open
Abstract
Traumatic brain injury (TBI) is characterized by complex secondary injury processes involving the p75 neurotrophin receptor (p75NTR), which has been proposed as a possible therapeutic target. However, the pathogenic role of the p75NTR co-receptor sortilin in TBI has not been investigated. In this study, we examined whether sortilin contributes to acute and early processes of secondary injury using a murine controlled cortical impact (CCI) model of TBI. Initial expression analysis showed a down-regulation of sortilin mRNA levels 1 and 5 day post injury (dpi) and a reduced expression of sortilin protein 1 dpi. Next, a total of 40 SortilinΔExon14 loss-of-function mouse mutants (Sort1-/-) and wild-type (Sort1+/+) littermate mice were subjected to CCI and examined at 1 and 5 dpi. Neither sensorimotor deficits or brain lesion size nor CCI-induced cell death or calcium-dependent excitotoxicity as evaluated by TUNEL staining or Western blot analysis of alpha II spectrin breakdown products were different between Sort1-/- and Sort1+/+ mice. In addition, CCI induced the up-regulation of pro-inflammatory marker mRNA expression (Il6, Tnfa, Aif1, and Gfap) irrespectively of the genotype. Similarly, the mRNA expressions of neurotrophins (Bdnf, Ngf, Nt3), VPS10P domain receptors others than sortilin (Ngfr, Sorl1, Sorcs2), and the sortilin interactor progranulin were not affected by genotype. Our results suggest that sortilin is a modulatory rather than a critical factor in the acute and early brain tissue response after TBI.
Collapse
Affiliation(s)
- Irina Staib-Lasarzik
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Christina Gölz
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Wieslawa Bobkiewiecz
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Pawit Somnuke
- Department of Anesthesiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Anne Sebastiani
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Serge C. Thal
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Michael K.E. Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neurosciences (FTN) of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
4
|
Chandra J. The potential role of the p75 receptor in schizophrenia: neuroimmunomodulation and making life or death decisions. Brain Behav Immun Health 2024; 38:100796. [PMID: 38813083 PMCID: PMC11134531 DOI: 10.1016/j.bbih.2024.100796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/06/2024] [Accepted: 05/12/2024] [Indexed: 05/31/2024] Open
Abstract
The nerve growth factor receptor, also referred to as tumour necrosis factor II and the p75 neurotrophin receptor (p75), serves pleiotropic functions in both the peripheral and central nervous system, involving modulation of immune responses, cell survival and cell death signalling in response to multiple ligands including cytokines such as TNFα, as well as proneurotrophins and mature neurotrophins. Whilst in vitro and in vivo studies have characterised various responses of the p75 receptor in isolated conditions, it remains unclear whether the p75 receptor serves to provide neuroprotection or contributes to neurotoxicity in neuroinflammatory and neurotrophin-deficit conditions, such as those presenting in schizophrenia. The purpose of this mini-review is to characterise the potential signalling mechanisms of the p75 receptor respective to neuropathological changes prevailing in schizophrenia to ultimately propose how specific functions of the receptor may underlie altered levels of p75 in specific cell types. On the basis of this evaluation, this mini-review aims to promote avenues for future research in utilising the therapeutic potential of ligands for the p75 receptor in psychiatric disorders, whereby heightened inflammation and reductions in trophic signalling mechanisms coalesce in the brain, potentially resulting in tissue damage.
Collapse
Affiliation(s)
- Jessica Chandra
- Neuroscience Research Australia, University of New South Wales, Sydney, Australia
| |
Collapse
|
5
|
Hubel E, Neumann A, Fishman S, Schaffer O, Erez N, Shrkihe BA, Shteingard Y, Gross T, Shibolet O, Varol C, Zvibel I. Sortilin in Biliary Epithelial Cells Promotes Ductular Reaction and Fibrosis during Cholestatic Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:941-957. [PMID: 38493927 DOI: 10.1016/j.ajpath.2024.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/03/2024] [Accepted: 01/29/2024] [Indexed: 03/19/2024]
Abstract
Cholestatic injuries are accompanied by ductular reaction, initiated by proliferation and activation of biliary epithelial cells (BECs), leading to fibrosis. Sortilin (encoded by Sort1) facilitates IL-6 secretion and leukemia inhibitory factor (LIF) signaling. This study investigated the interplay between sortilin and IL-6 and LIF in cholestatic injury-induced ductular reaction, morphogenesis of new ducts, and fibrosis. Cholestatic injury was induced by bile duct ligation (BDL) in wild-type and Sort1-/- mice, with or without augmentation of IL-6 or LIF. Mice with BEC sortilin deficiency (hGFAPcre.Sort1fl/fl) and control mice were subjected to BDL and 3,5-diethoxycarbonyl-1,4-dihydrocollidine diet (DDC) induced cholestatic injury. Sort1-/- mice displayed reduced BEC proliferation and expression of BEC-reactive markers. Administration of LIF or IL-6 restored BEC proliferation in Sort1-/- mice, without affecting BEC-reactive or inflammatory markers. Sort1-/- mice also displayed impaired morphogenesis, which was corrected by LIF treatment. Similarly, hGFAPcre.Sort1fl/fl mice exhibited reduced BEC proliferation, but similar reactive and inflammatory marker expression. Serum IL-6 and LIF were comparable, yet liver pSTAT3 was reduced, indicating that sortilin is essential for co-activation of LIF receptor/gp130 signaling in BECs, but not for IL-6 secretion. hGFAPcre.Sortfl/fl mice displayed impaired morphogenesis and diminished fibrosis after BDL and DDC. In conclusion, sortilin-mediated engagement of LIF signaling in BECs promoted ductular reaction and morphogenesis during cholestatic injury. This study indicates that BEC sortilin is pivotal for the development of fibrosis.
Collapse
Affiliation(s)
- Einav Hubel
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Anat Neumann
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Sigal Fishman
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ortal Schaffer
- Department of Pediatric Surgery, Assaf Harofe Hospital, Tzrifin, Israel
| | - Noam Erez
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Bander Abu Shrkihe
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yuval Shteingard
- Department of Pathology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Tamar Gross
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Oren Shibolet
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Chen Varol
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Clinical Microbiology and Immunology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| | - Isabel Zvibel
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
6
|
Zhao Y, Yang L, Que S, An L, Teeti AA, Xiao S. Systemic mechanism of Panax noteginseng saponins in antiaging based on network pharmacology combined with experimental validation. IBRAIN 2024; 10:519-535. [PMID: 39691419 PMCID: PMC11649391 DOI: 10.1002/ibra.12165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 12/19/2024]
Abstract
This study aims to investigate the systemic mechanism of Panax notoginseng saponins (PNS) in antiaging using network pharmacology combined with experimental validation. String database and Cytoscape3.7.2 were used to perform the protein-protein interaction (PPI) and construct genes network. The key target genes were analyzed using gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Then, the aging-related genes were verified by reverse-transcription polymerase chain reaction in SAM-P/8 mice, and performed molecular docking with the main components of PNS. Moreover, it produced cluster between Hub genes and differential genes. A total of 169 crossover genes were obtained, and the results of GO and KEGG indicated that the antiaging effect of PNS was mediated by apoptosis, cancer, and neurodegeneration and that five of the eight Hub genes had good binding activity with the main components of PNS. In addition, animal experiments reported that MAP2, MAPKK4, RAB6A, and Sortilin-1 have different levels of expression in the brain tissues of aging mice, and bind well docking with the main active components of PNS. However, there was no crossover between the 169 PNS intersecting genes and the four differential genes, while they yielded a link from PPI in which MAP2K4 was only linked to AKT1 and CASP3; MAP2 was only linked to AKT1 and CASP3; RAB6A was only linked to AKT1; but Sortlin-1 did not link to the Hub genes. In summary, the antiaging effect of PNS is associated with the eight Hub genes and four differential genes. All of them consist of a cluster or group that is possibly related to the antiaging effect of PNS.
Collapse
Affiliation(s)
- Yang‐Yang Zhao
- Department of NeurosurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Li‐Xia Yang
- Department of NeurosurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Shuang‐Yu Que
- Department of NeurosurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Lei‐Xing An
- Department of NeurosurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Abeer A. Teeti
- Department of Chemistry, School of ScienceHebron UniversityHebronPalestine
| | - Shun‐Wu Xiao
- Department of NeurosurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| |
Collapse
|
7
|
Li Z, Liu D, Wang G, Zheng Y, Chen L, Cheng Z, Zhang Z, Cai Q, Ge F, Fan Y, Guan X. METH exposure alters sperm DNA methylation in F0 mice and mPFC transcriptome in male F1 mice. Psychopharmacology (Berl) 2024; 241:897-911. [PMID: 38092953 DOI: 10.1007/s00213-023-06516-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/29/2023] [Indexed: 04/20/2024]
Abstract
RATIONALE Methamphetamine (METH) exposure has toxicity in sperm epigenetic phenotype and increases the risk for developing addiction in their offspring. However, the underlying transgenerational mechanism remains unclear. OBJECTIVES The current study aims to investigate the profiles of sperm epigenetic modifications in male METH-exposed mice (F0) and medial prefrontal cortex (mPFC) transcriptome in their male first-generation offspring (F1). METHODS METH-related male F0 and F1 mice model was established to investigate the effects of paternal METH exposure on reproductive functions and sperm DNA methylation in F0 and mPFC transcriptomic profile in F1. During adulthood, F1 was subjected to a conditioned place preference (CPP) test to evaluate sensitivity to METH. The gene levels were verified with qPCR. RESULTS METH exposure obviously altered F0 sperms DNA methylated profile and male F1 mPFC transcriptomic profile, many of which being related to neuronal system and brain development. In METH-sired male F1, subthreshold dose of METH administration effectively elicited CPP, along with more mPFC activation. After qPCR verification, Sort1 and Shank2 were at higher levels in F0 sperm and F1 mPFC. CONCLUSIONS Our findings put new insights into paternal METH exposure-altered profiles of F0 sperm DNA methylation and male F1 mPFC transcriptomics. Several genes, such as Sort1 and Shank2, might be used as potential molecules for further research on the transgenerational vulnerability to drug addiction in offspring by paternal drug exposure.
Collapse
Affiliation(s)
- Zhaosu Li
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Dekang Liu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guanxiong Wang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yanyan Zheng
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Liying Chen
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhen Cheng
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zijing Zhang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qinglong Cai
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Feifei Ge
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yu Fan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiaowei Guan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
8
|
Thomasen PB, Salasova A, Kjaer-Sorensen K, Woloszczuková L, Lavický J, Login H, Tranberg-Jensen J, Almeida S, Beel S, Kavková M, Qvist P, Kjolby M, Ovesen PL, Nolte S, Vestergaard B, Udrea AC, Nejsum LN, Chao MV, Van Damme P, Krivanek J, Dasen J, Oxvig C, Nykjaer A. SorCS2 binds progranulin to regulate motor neuron development. Cell Rep 2023; 42:113333. [PMID: 37897724 DOI: 10.1016/j.celrep.2023.113333] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/25/2023] [Accepted: 10/09/2023] [Indexed: 10/30/2023] Open
Abstract
Motor neuron (MN) development and nerve regeneration requires orchestrated action of a vast number of molecules. Here, we identify SorCS2 as a progranulin (PGRN) receptor that is required for MN diversification and axon outgrowth in zebrafish and mice. In zebrafish, SorCS2 knockdown also affects neuromuscular junction morphology and fish motility. In mice, SorCS2 and PGRN are co-expressed by newborn MNs from embryonic day 9.5 until adulthood. Using cell-fate tracing and nerve segmentation, we find that SorCS2 deficiency perturbs cell-fate decisions of brachial MNs accompanied by innervation deficits of posterior nerves. Additionally, adult SorCS2 knockout mice display slower motor nerve regeneration. Interestingly, primitive macrophages express high levels of PGRN, and their interaction with SorCS2-positive motor axon is required during axon pathfinding. We further show that SorCS2 binds PGRN to control its secretion, signaling, and conversion into granulins. We propose that PGRN-SorCS2 signaling controls MN development and regeneration in vertebrates.
Collapse
Affiliation(s)
- Pernille Bogetofte Thomasen
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Alena Salasova
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.
| | - Kasper Kjaer-Sorensen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Lucie Woloszczuková
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Josef Lavický
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Hande Login
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Jeppe Tranberg-Jensen
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Sergio Almeida
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Sander Beel
- Department of Neurology and Department of Neurosciences, KU Leuven and Center for Brain & Disease Research VIB, 3000 Leuven, Belgium
| | - Michaela Kavková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Per Qvist
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Mads Kjolby
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Peter Lund Ovesen
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Stella Nolte
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Benedicte Vestergaard
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Andreea-Cornelia Udrea
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | | | - Moses V Chao
- Department of Neuroscience and Physiology, NYU Langone Health, New York, NY 10016, USA
| | - Philip Van Damme
- Department of Neurology and Department of Neurosciences, KU Leuven and Center for Brain & Disease Research VIB, 3000 Leuven, Belgium
| | - Jan Krivanek
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Jeremy Dasen
- Department of Neuroscience and Physiology, NYU Langone Health, New York, NY 10016, USA
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Anders Nykjaer
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.
| |
Collapse
|
9
|
Kurnellas M, Mitra A, Schwabe T, Paul R, Arrant AE, Roberson ED, Ward M, Yeh F, Long H, Rosenthal A. Latozinemab, a novel progranulin-elevating therapy for frontotemporal dementia. J Transl Med 2023; 21:387. [PMID: 37322482 PMCID: PMC10268535 DOI: 10.1186/s12967-023-04251-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 06/08/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Heterozygous loss-of-function mutations in the progranulin (PGRN) gene (GRN) cause a reduction in PGRN and lead to the development of frontotemporal dementia (FTD-GRN). PGRN is a secreted lysosomal chaperone, immune regulator, and neuronal survival factor that is shuttled to the lysosome through multiple receptors, including sortilin. Here, we report the characterization of latozinemab, a human monoclonal antibody that decreases the levels of sortilin, which is expressed on myeloid and neuronal cells and shuttles PGRN to the lysosome for degradation, and blocks its interaction with PGRN. METHODS In vitro characterization studies were first performed to assess the mechanism of action of latozinemab. After the in vitro studies, a series of in vivo studies were performed to assess the efficacy of a mouse-cross reactive anti-sortilin antibody and the pharmacokinetics, pharmacodynamics, and safety of latozinemab in nonhuman primates and humans. RESULTS In a mouse model of FTD-GRN, the rodent cross-reactive anti-sortilin antibody, S15JG, decreased total sortilin levels in white blood cell (WBC) lysates, restored PGRN to normal levels in plasma, and rescued a behavioral deficit. In cynomolgus monkeys, latozinemab decreased sortilin levels in WBCs and concomitantly increased plasma and cerebrospinal fluid (CSF) PGRN by 2- to threefold. Finally, in a first-in-human phase 1 clinical trial, a single infusion of latozinemab caused a reduction in WBC sortilin, tripled plasma PGRN and doubled CSF PGRN in healthy volunteers, and restored PGRN to physiological levels in asymptomatic GRN mutation carriers. CONCLUSIONS These findings support the development of latozinemab for the treatment of FTD-GRN and other neurodegenerative diseases where elevation of PGRN may be beneficial. Trial registration ClinicalTrials.gov, NCT03636204. Registered on 17 August 2018, https://clinicaltrials.gov/ct2/show/NCT03636204 .
Collapse
Affiliation(s)
- Michael Kurnellas
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA.
- Neuron23, South San Francisco, CA, 94080, USA.
| | - Ananya Mitra
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
| | - Tina Schwabe
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
- Nine Square Therapeutics, Inc., South San Francisco, CA, 94080, USA
| | - Robert Paul
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
- Nine Square Therapeutics, Inc., South San Francisco, CA, 94080, USA
| | - Andrew E Arrant
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, and Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Erik D Roberson
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, and Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael Ward
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
| | - Felix Yeh
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
- Genentech, South San Francisco, CA, 94080, USA
| | - Hua Long
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
| | - Arnon Rosenthal
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
| |
Collapse
|
10
|
Milyutina YP, Arutjunyan AV, Korenevsky AV, Selkov SA, Kogan IY. Neurotrophins: are they involved in immune tolerance in pregnancy? Am J Reprod Immunol 2023; 89:e13694. [PMID: 36792972 DOI: 10.1111/aji.13694] [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: 09/29/2022] [Revised: 01/06/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
In this review, an attempt was made to substantiate the possibility for neurotrophins to be involved in the development of immune tolerance based on data accumulated on neurotrophin content and receptor expression in the trophoblast and immune cells, in particular, in natural killer cells. Numerous research results are reviewed to show that the expression and localization of neurotrophins along with their high-affinity tyrosine kinase receptors and low-affinity p75NTR receptor in the mother-placenta-fetus system indicate the important role of neurotrophins as binding molecules in regulating the crosstalk between the nervous, endocrine, and immune systems in pregnancy. An imbalance between these systems can occur with tumor growth and pathological processes observed in pregnancy complications and fetal development anomalies.
Collapse
Affiliation(s)
- Yulia P Milyutina
- D.O. Ott Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, Russia
- St. Petersburg State Pediatric Medical University, St. Petersburg, Russia
| | - Alexander V Arutjunyan
- D.O. Ott Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, Russia
| | - Andrey V Korenevsky
- D.O. Ott Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, Russia
| | - Sergey A Selkov
- D.O. Ott Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, Russia
| | - Igor Yu Kogan
- D.O. Ott Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, Russia
| |
Collapse
|
11
|
Elia A, Fossati S. Autonomic nervous system and cardiac neuro-signaling pathway modulation in cardiovascular disorders and Alzheimer's disease. Front Physiol 2023; 14:1060666. [PMID: 36798942 PMCID: PMC9926972 DOI: 10.3389/fphys.2023.1060666] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/19/2023] [Indexed: 01/31/2023] Open
Abstract
The heart is a functional syncytium controlled by a delicate and sophisticated balance ensured by the tight coordination of its several cell subpopulations. Accordingly, cardiomyocytes together with the surrounding microenvironment participate in the heart tissue homeostasis. In the right atrium, the sinoatrial nodal cells regulate the cardiac impulse propagation through cardiomyocytes, thus ensuring the maintenance of the electric network in the heart tissue. Notably, the central nervous system (CNS) modulates the cardiac rhythm through the two limbs of the autonomic nervous system (ANS): the parasympathetic and sympathetic compartments. The autonomic nervous system exerts non-voluntary effects on different peripheral organs. The main neuromodulator of the Sympathetic Nervous System (SNS) is norepinephrine, while the principal neurotransmitter of the Parasympathetic Nervous System (PNS) is acetylcholine. Through these two main neurohormones, the ANS can gradually regulate cardiac, vascular, visceral, and glandular functions by turning on one of its two branches (adrenergic and/or cholinergic), which exert opposite effects on targeted organs. Besides these neuromodulators, the cardiac nervous system is ruled by specific neuropeptides (neurotrophic factors) that help to preserve innervation homeostasis through the myocardial layers (from epicardium to endocardium). Interestingly, the dysregulation of this neuro-signaling pathway may expose the cardiac tissue to severe disorders of different etiology and nature. Specifically, a maladaptive remodeling of the cardiac nervous system may culminate in a progressive loss of neurotrophins, thus leading to severe myocardial denervation, as observed in different cardiometabolic and neurodegenerative diseases (myocardial infarction, heart failure, Alzheimer's disease). This review analyzes the current knowledge on the pathophysiological processes involved in cardiac nervous system impairment from the perspectives of both cardiac disorders and a widely diffused and devastating neurodegenerative disorder, Alzheimer's disease, proposing a relationship between neurodegeneration, loss of neurotrophic factors, and cardiac nervous system impairment. This overview is conducive to a more comprehensive understanding of the process of cardiac neuro-signaling dysfunction, while bringing to light potential therapeutic scenarios to correct or delay the adverse cardiovascular remodeling, thus improving the cardiac prognosis and quality of life in patients with heart or neurodegenerative disorders.
Collapse
|
12
|
Honfozo A, Ghouil R, Alayi TD, Ouldali M, Arteni AA, Atindehou CM, Fanou LA, Hathout Y, Zinn-Justin S, Tomavo S. The luminal domain of Toxoplasma gondii sortilin adopts a ring-shaped structure exhibiting motifs specific to apicomplexan parasites. FRONTIERS IN PARASITOLOGY 2023; 2:1103772. [PMID: 39816831 PMCID: PMC11731604 DOI: 10.3389/fpara.2023.1103772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/17/2023] [Indexed: 01/18/2025]
Abstract
Rhoptries and micronemes are essential for host cell invasion and survival of all apicomplexan parasites, which are composed of numerous obligate intracellular protozoan pathogens including Plasmodium falciparum (malaria) and Toxoplasma gondii (toxoplasmosis) that infect humans and animals causing severe diseases. We identified Toxoplasma gondii TgSORT as an essential cargo receptor, which drives the transport of rhoptry (ROP) and microneme (MIC) proteins to ensure the biogenesis of these secretory organelles. The luminal domain of 752 amino acid long situated at the N-terminus end of TgSORT has been described to bind to MIC and ROP proteins. Here, we present an optimized protocol for expression of the entire luminal N-terminus of TgSORT (Tg-NSORT) in the yeast Pichia pastoris. Optimization of its coding sequence, cloning and transformation of the yeast P. pastoris allowed the secretion of Tg-NSORT. The protein was purified and further analyzed by negative staining electron microscopy. In addition, molecular modeling using AlphaFold identified key differences between the human and the T gondii sortilin. The structural features that are only present in T. gondii and other apicomplexan parasites were highlighted. Elucidating the roles of these specific structural features may be useful for designing new therapeutic agents against apicomplexan parasites.
Collapse
Affiliation(s)
- Ariane Honfozo
- Université Paris Saclay, CNRS UMR 9198-CEA, Institute for Integrative Biology of the Cell (I2BC), Gif sur Yvette, France
| | - Rania Ghouil
- Université Paris Saclay, CNRS UMR 9198-CEA, Institute for Integrative Biology of the Cell (I2BC), Gif sur Yvette, France
| | | | - Malika Ouldali
- Université Paris Saclay, CNRS UMR 9198-CEA, Institute for Integrative Biology of the Cell (I2BC), Gif sur Yvette, France
| | - Ana-Andreea Arteni
- Université Paris Saclay, CNRS UMR 9198-CEA, Institute for Integrative Biology of the Cell (I2BC), Gif sur Yvette, France
| | - Cynthia Menonve Atindehou
- Laboratoire de Biochimie et de Biologie Moléculaire, Faculté des Sciences et Technologies, Université d’Abomey Calavi, Cotonou, Benin
| | - Lucie Ayi Fanou
- Laboratoire de Biochimie et de Biologie Moléculaire, Faculté des Sciences et Technologies, Université d’Abomey Calavi, Cotonou, Benin
| | - Yetrib Hathout
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, United States
| | - Sophie Zinn-Justin
- Université Paris Saclay, CNRS UMR 9198-CEA, Institute for Integrative Biology of the Cell (I2BC), Gif sur Yvette, France
| | - Stanislas Tomavo
- Université Paris Saclay, CNRS UMR 9198-CEA, Institute for Integrative Biology of the Cell (I2BC), Gif sur Yvette, France
| |
Collapse
|
13
|
Chen Y, Zhu XW, Lai WF, Liu YP, Xu XF, Liu LM, Chen YJ, Zhang CF, Wang GY, Cheng ZQ, Liu DZ. Gancao Nourishing-Yin decoction combined with methotrexate in treatment of aging CIA mice: a study based on DIA proteomic analysis. Chin Med 2023; 18:9. [PMID: 36709303 PMCID: PMC9883815 DOI: 10.1186/s13020-023-00709-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/04/2023] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Elderly rheumatoid arthritis (ERA) population faces multiple treatment dilemma. Here we aim to investigate if Gancao Nourishing-Yin decoction (GCNY) added to methotrexate (MTX) exhibit better effects in an ERA mice model. METHODS ERA mice model was established by adding D-galactose (Dgal) to collagen-induced arthritis (CIA) mice. The model was then assigned into control group (CIA + Dgal), MTX treatment group (MTX), GCNY treatment group (GCNY), and integrative treatment group (MTX + GCNY). Pathological scoring was performed to evaluate the severity between the groups. Proteomic analysis was applied to investigate the secretory phenotype of the ERA mouse model and the underlying mechanism of GCNY, MTX and their combination. Representative cytokines related to proteomic results were further validated by ELISAs. RESULTS CIA + Dgal mice showed more aggressive joints damage than the CIA mice. Besides changes in the inflammatory pathway such as Pi3k-Akt signaling pathway in both model, differential expressed proteins (DEPs) indicated metabolism-related pathways were more obvious in CIA + Dgal mice. Low-dose MTX failed to show pathological improvement in CIA + Dgal mice, while GCNY improved joints damage significantly. Besides down-regulated inflammation-related targets, GCNY-regulated DEPs (such as Apoc1 ~ 3, Grk2 and Creb3l3) were broadly enriched in metabolism-related pathways. MTX + GCNY showed the best therapeutic effect, and the DEPs enriched in a variety of inflammatory,metabolism and osteoclast differentiation signaling pathway. Notably, MTX + GCNY treatment up-regulated Dhfr, Cbr1, Shmt1 involved in folic acid biosynthesis and anti-folate resistance pathways indicated a coincidence synergic action. ELISAs confirmed CPR and Akt that elevated in CIA + Dgal mice were significantly ameliorated by treatments, and adding on GCNY elevated folic acid levels and its regulator Dhfr. CONCLUSION Aging aggravated joints damage in CIA, which probably due to metabolic changes rather than more severe inflammation. GCNY showed significant effects in the ERA mice model especially when integrated with MTX to obtain a synergic action.
Collapse
Affiliation(s)
- Yong Chen
- grid.440218.b0000 0004 1759 7210Shenzhen People‘s Hospital (The Second Clinical Medical College of Jinan University and the First Affiliated Hospital to Southern University of Science and Technology), Shenzhen, China ,grid.440218.b0000 0004 1759 7210Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People‘s Hospital, Shenzhen, China
| | - Xing-wang Zhu
- grid.284723.80000 0000 8877 7471Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou, China
| | - Wing-Fu Lai
- grid.16890.360000 0004 1764 6123Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong, Hong Kong Special Administrative Region China ,Department of Urology, Zhejiang Provincial People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang China
| | - Yong-pu Liu
- grid.284723.80000 0000 8877 7471Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou, China
| | - Xuan-feng Xu
- grid.284723.80000 0000 8877 7471Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou, China
| | - Li-ming Liu
- grid.440218.b0000 0004 1759 7210Shenzhen People‘s Hospital (The Second Clinical Medical College of Jinan University and the First Affiliated Hospital to Southern University of Science and Technology), Shenzhen, China
| | - Yan-juan Chen
- grid.440218.b0000 0004 1759 7210Shenzhen People‘s Hospital (The Second Clinical Medical College of Jinan University and the First Affiliated Hospital to Southern University of Science and Technology), Shenzhen, China
| | - Chuan-fu Zhang
- grid.412540.60000 0001 2372 7462The Seventh People’s Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang-yi Wang
- grid.413458.f0000 0000 9330 9891Guizhou Medical University affiliated Hospital, Guiyang, China
| | - Zhi-qiang Cheng
- grid.440218.b0000 0004 1759 7210Shenzhen People‘s Hospital (The Second Clinical Medical College of Jinan University and the First Affiliated Hospital to Southern University of Science and Technology), Shenzhen, China
| | - Dong-zhou Liu
- grid.440218.b0000 0004 1759 7210Shenzhen People‘s Hospital (The Second Clinical Medical College of Jinan University and the First Affiliated Hospital to Southern University of Science and Technology), Shenzhen, China
| |
Collapse
|
14
|
Salasova A, Monti G, Andersen OM, Nykjaer A. Finding memo: versatile interactions of the VPS10p-Domain receptors in Alzheimer’s disease. Mol Neurodegener 2022; 17:74. [PMID: 36397124 PMCID: PMC9673319 DOI: 10.1186/s13024-022-00576-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 10/17/2022] [Indexed: 11/19/2022] Open
Abstract
The family of VPS10p-Domain (D) receptors comprises five members named SorLA, Sortilin, SorCS1, SorCS2 and SorCS3. While their physiological roles remain incompletely resolved, they have been recognized for their signaling engagements and trafficking abilities, navigating a number of molecules between endosome, Golgi compartments, and the cell surface. Strikingly, recent studies connected all the VPS10p-D receptors to Alzheimer’s disease (AD) development. In addition, they have been also associated with diseases comorbid with AD such as diabetes mellitus and major depressive disorder. This systematic review elaborates on genetic, functional, and mechanistic insights into how dysfunction in VPS10p-D receptors may contribute to AD etiology, AD onset diversity, and AD comorbidities. Starting with their functions in controlling cellular trafficking of amyloid precursor protein and the metabolism of the amyloid beta peptide, we present and exemplify how these receptors, despite being structurally similar, regulate various and distinct cellular events involved in AD. This includes a plethora of signaling crosstalks that impact on neuronal survival, neuronal wiring, neuronal polarity, and synaptic plasticity. Signaling activities of the VPS10p-D receptors are especially linked, but not limited to, the regulation of neuronal fitness and apoptosis via their physical interaction with pro- and mature neurotrophins and their receptors. By compiling the functional versatility of VPS10p-D receptors and their interactions with AD-related pathways, we aim to further propel the AD research towards VPS10p-D receptor family, knowledge that may lead to new diagnostic markers and therapeutic strategies for AD patients.
Collapse
|
15
|
Xiong LL, Chen L, Deng IB, Zhou XF, Wang TH. P75 neurotrophin receptor as a therapeutic target for drug development to treat neurological diseases. Eur J Neurosci 2022; 56:5299-5318. [PMID: 36017737 DOI: 10.1111/ejn.15810] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 12/14/2022]
Abstract
The interaction of neurotrophins with their receptors is involved in the pathogenesis and progression of various neurological diseases, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, spinal cord injury and acute and chronic cerebral damage. The p75 neurotrophin receptor (p75NTR) plays a pivotal role in the development of neurological dysfunctions as a result of its high expression, abnormal processing and signalling. Therefore, p75NTR represents as a vital therapeutic target for the treatment of neurodegeneration, neuropsychiatric disorders and cerebrovascular insufficiency. This review summarizes the current research progress on the p75NTR signalling in neurological deficits. We also summarize the present therapeutic approaches by genetically and pharmacologically targeting p75NTR for the attenuation of pathological changes. Based on the evolving knowledge, the role of p75NTR in the regulation of tau hyperphosphorylation, Aβ metabolism, the degeneration of motor neurons and dopaminergic neurons has been discussed. Its position as a biomarker to evaluate the severity of diseases and as a druggable target for drug development has also been elucidated. Several prototype small molecule compounds were introduced to be crucial in neuronal survival and functional recovery via targeting p75NTR. These small molecule compounds represent desirable agents in attenuating neurodegeneration and cell death as they abolish activation-induced neurotoxicity of neurotrophins via modulating p75NTR signalling. More comprehensive and in-depth investigations on p75NTR-based drug development are required to shed light on effective treatment of numerous neurological disorders.
Collapse
Affiliation(s)
- Liu-Lin Xiong
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, China.,Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia.,Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Li Chen
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Isaac Bul Deng
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Xin-Fu Zhou
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Ting-Hua Wang
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Zhang Y, Gao X, Bai X, Yao S, Chang YZ, Gao G. The emerging role of furin in neurodegenerative and neuropsychiatric diseases. Transl Neurodegener 2022; 11:39. [PMID: 35996194 PMCID: PMC9395820 DOI: 10.1186/s40035-022-00313-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/10/2022] [Indexed: 12/02/2022] Open
Abstract
Furin is an important mammalian proprotein convertase that catalyzes the proteolytic maturation of a variety of prohormones and proproteins in the secretory pathway. In the brain, the substrates of furin include the proproteins of growth factors, receptors and enzymes. Emerging evidence, such as reduced FURIN mRNA expression in the brains of Alzheimer's disease patients or schizophrenia patients, has implicated a crucial role of furin in the pathophysiology of neurodegenerative and neuropsychiatric diseases. Currently, compared to cancer and infectious diseases, the aberrant expression of furin and its pharmaceutical potentials in neurological diseases remain poorly understood. In this article, we provide an overview on the physiological roles of furin and its substrates in the brain, summarize the deregulation of furin expression and its effects in neurodegenerative and neuropsychiatric disorders, and discuss the implications and current approaches that target furin for therapeutic interventions. This review may expedite future studies to clarify the molecular mechanisms of furin deregulation and involvement in the pathogenesis of neurodegenerative and neuropsychiatric diseases, and to develop new diagnosis and treatment strategies for these diseases.
Collapse
Affiliation(s)
- Yi Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Xiaoqin Gao
- Shijiazhuang People's Hospital, Hebei Medical University, Shijiazhuang, 050027, China
| | - Xue Bai
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Shanshan Yao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Yan-Zhong Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| | - Guofen Gao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| |
Collapse
|
17
|
Mitok KA, Keller MP, Attie AD. Sorting through the extensive and confusing roles of sortilin in metabolic disease. J Lipid Res 2022; 63:100243. [PMID: 35724703 PMCID: PMC9356209 DOI: 10.1016/j.jlr.2022.100243] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 01/06/2023] Open
Abstract
Sortilin is a post-Golgi trafficking receptor homologous to the yeast vacuolar protein sorting receptor 10 (VPS10). The VPS10 motif on sortilin is a 10-bladed β-propeller structure capable of binding more than 50 proteins, covering a wide range of biological functions including lipid and lipoprotein metabolism, neuronal growth and death, inflammation, and lysosomal degradation. Sortilin has a complex cellular trafficking itinerary, where it functions as a receptor in the trans-Golgi network, endosomes, secretory vesicles, multivesicular bodies, and at the cell surface. In addition, sortilin is associated with hypercholesterolemia, Alzheimer's disease, prion diseases, Parkinson's disease, and inflammation syndromes. The 1p13.3 locus containing SORT1, the gene encoding sortilin, carries the strongest association with LDL-C of all loci in human genome-wide association studies. However, the mechanism by which sortilin influences LDL-C is unclear. Here, we review the role sortilin plays in cardiovascular and metabolic diseases and describe in detail the large and often contradictory literature on the role of sortilin in the regulation of LDL-C levels.
Collapse
Affiliation(s)
- Kelly A Mitok
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Alan D Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
18
|
Mehterov N, Minchev D, Gevezova M, Sarafian V, Maes M. Interactions Among Brain-Derived Neurotrophic Factor and Neuroimmune Pathways Are Key Components of the Major Psychiatric Disorders. Mol Neurobiol 2022; 59:4926-4952. [PMID: 35657457 DOI: 10.1007/s12035-022-02889-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/17/2022] [Indexed: 10/25/2022]
Abstract
The purpose of this review is to summarize the current knowledge regarding the reciprocal associations between brain-derived neurotrophic factor (BDNF) and immune-inflammatory pathways and how these links may explain the involvement of this neurotrophin in the immune pathophysiology of mood disorders and schizophrenia. Toward this end, we delineated the protein-protein interaction (PPI) network centered around BDNF and searched PubMed, Scopus, Google Scholar, and Science Direct for papers dealing with the involvement of BDNF in the major psychosis, neurodevelopment, neuronal functions, and immune-inflammatory and related pathways. The PPI network was built based on the significant interactions of BDNF with neurotrophic (NTRK2, NTF4, and NGFR), immune (cytokines, STAT3, TRAF6), and cell-cell junction (CTNNB, CDH1) DEPs (differentially expressed proteins). Enrichment analysis shows that the most significant terms associated with this PPI network are the tyrosine kinase receptor (TRKR) and Src homology region two domain-containing phosphatase-2 (SHP2) pathways, tyrosine kinase receptor signaling pathways, positive regulation of kinase and transferase activity, cytokine signaling, and negative regulation of the immune response. The participation of BDNF in the immune response and its interactions with neuroprotective and cell-cell adhesion DEPs is probably a conserved regulatory process which protects against the many detrimental effects of immune activation and hyperinflammation including neurotoxicity. Lowered BDNF levels in mood disorders and schizophrenia (a) are associated with disruptions in neurotrophic signaling and activated immune-inflammatory pathways leading to neurotoxicity and (b) may interact with the reduced expression of other DEPs (CTNNB1, CDH1, or DISC1) leading to multiple aberrations in synapse and axonal functions.
Collapse
Affiliation(s)
- Nikolay Mehterov
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria.,Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Danail Minchev
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria.,Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Maria Gevezova
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria.,Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Victoria Sarafian
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria.,Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Michael Maes
- Faculty of Medicine, Department of Psychiatry, Chulalongkorn University, Bangkok, 10330, Thailand. .,Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria. .,Department of Psychiatry, IMPACT Strategic Research Centre, Deakin University, Geelong, VIC, Australia.
| |
Collapse
|
19
|
Ulrichsen M, Gonçalves NP, Mohseni S, Hjæresen S, Lisle TL, Molgaard S, Madsen NK, Andersen OM, Svenningsen ÅF, Glerup S, Nykjær A, Vægter CB. Sortilin Modulates Schwann Cell Signaling and Remak Bundle Regeneration Following Nerve Injury. Front Cell Neurosci 2022; 16:856734. [PMID: 35634462 PMCID: PMC9130554 DOI: 10.3389/fncel.2022.856734] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Peripheral nerve regeneration relies on the ability of Schwann cells to support the regrowth of damaged axons. Schwann cells re-differentiate when reestablishing contact with the sprouting axons, with large fibers becoming remyelinated and small nociceptive fibers ensheathed and collected into Remak bundles. We have previously described how the receptor sortilin facilitates neurotrophin signaling in peripheral neurons via regulated trafficking of Trk receptors. This study aims to characterize the effects of sortilin deletion on nerve regeneration following sciatic crush injury. We found that Sort1–/– mice displayed functional motor recovery like that of WT mice, with no detectable differences in relation to nerve conduction velocities and morphological aspects of myelinated fibers. In contrast, we found abnormal ensheathment of regenerated C-fibers in injured Sort1–/– mice, demonstrating a role of sortilin for Remak bundle formation following injury. Further studies on Schwann cell signaling pathways showed a significant reduction of MAPK/ERK, RSK, and CREB phosphorylation in Sort1–/– Schwann cells after stimulation with neurotrophin-3 (NT-3), while Schwann cell migration and myelination remained unaffected. In conclusion, our results demonstrate that loss of sortilin blunts NT-3 signaling in Schwann cells which might contribute to the impaired Remak bundle regeneration after sciatic nerve injury.
Collapse
Affiliation(s)
- Maj Ulrichsen
- Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Nádia P. Gonçalves
- Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Simin Mohseni
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Simone Hjæresen
- Neurobiological Research, Faculty of Health Sciences, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Thomas L. Lisle
- Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Simon Molgaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Niels K. Madsen
- Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Olav M. Andersen
- Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Åsa F. Svenningsen
- Neurobiological Research, Faculty of Health Sciences, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Anders Nykjær
- Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
- Center of Excellence PROMEMO, Aarhus University, Aarhus, Denmark
| | - Christian B. Vægter
- Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- *Correspondence: Christian B. Vægter,
| |
Collapse
|
20
|
Conlon DM, Schneider CV, Ko YA, Rodrigues A, Guo K, Hand NJ, Rader DJ. Sortilin restricts secretion of apolipoprotein B-100 by hepatocytes under stressed but not basal conditions. J Clin Invest 2022; 132:144334. [PMID: 35113816 PMCID: PMC8920325 DOI: 10.1172/jci144334] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/02/2022] [Indexed: 12/02/2022] Open
Abstract
Genetic variants at the SORT1 locus in humans, which cause increased SORT1 expression in the liver, are significantly associated with reduced plasma levels of LDL cholesterol and apolipoprotein B (apoB). However, the role of hepatic sortilin remains controversial, as genetic deletion of sortilin in mice has resulted in variable and conflicting effects on apoB secretion. Here, we found that Sort1-KO mice on a chow diet and several Sort1-deficient hepatocyte lines displayed no difference in apoB secretion. When these models were challenged with high-fat diet or ER stress, the loss of Sort1 expression resulted in a significant increase in apoB-100 secretion. Sort1-overexpression studies yielded reciprocal results. Importantly, carriers of SORT1 variant with diabetes had larger decreases in plasma apoB, TG, and VLDL and LDL particle number as compared with people without diabetes with the same variants. We conclude that, under basal nonstressed conditions, loss of sortilin has little effect on hepatocyte apoB secretion, whereas, in the setting of lipid loading or ER stress, sortilin deficiency leads to increased apoB secretion. These results are consistent with the directionality of effect in human genetics studies and suggest that, under stress conditions, hepatic sortilin directs apoB toward lysosomal degradation rather than secretion, potentially serving as a quality control step in the apoB secretion pathway in hepatocytes.
Collapse
Affiliation(s)
- Donna M Conlon
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Carolin V Schneider
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Yi-An Ko
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Amrith Rodrigues
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Kathy Guo
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Nicholas J Hand
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Daniel J Rader
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| |
Collapse
|
21
|
Eggert S, Kins S, Endres K, Brigadski T. Brothers in arms: proBDNF/BDNF and sAPPα/Aβ-signaling and their common interplay with ADAM10, TrkB, p75NTR, sortilin, and sorLA in the progression of Alzheimer's disease. Biol Chem 2022; 403:43-71. [PMID: 34619027 DOI: 10.1515/hsz-2021-0330] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/16/2021] [Indexed: 12/22/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is an important modulator for a variety of functions in the central nervous system (CNS). A wealth of evidence, such as reduced mRNA and protein level in the brain, cerebrospinal fluid (CSF), and blood samples of Alzheimer's disease (AD) patients implicates a crucial role of BDNF in the progression of this disease. Especially, processing and subcellular localization of BDNF and its receptors TrkB and p75 are critical determinants for survival and death in neuronal cells. Similarly, the amyloid precursor protein (APP), a key player in Alzheimer's disease, and its cleavage fragments sAPPα and Aβ are known for their respective roles in neuroprotection and neuronal death. Common features of APP- and BDNF-signaling indicate a causal relationship in their mode of action. However, the interconnections of APP- and BDNF-signaling are not well understood. Therefore, we here discuss dimerization properties, localization, processing by α- and γ-secretase, relevance of the common interaction partners TrkB, p75, sorLA, and sortilin as well as shared signaling pathways of BDNF and sAPPα.
Collapse
Affiliation(s)
- Simone Eggert
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, D-67663 Kaiserslautern, Germany
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, D-67663 Kaiserslautern, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany
| | - Tanja Brigadski
- Department of Informatics and Microsystem Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibrücken, Germany
| |
Collapse
|
22
|
Asaro A, Sinha R, Bakun M, Kalnytska O, Carlo-Spiewok AS, Rubel T, Rozeboom A, Dadlez M, Kaminska B, Aronica E, Malik AR, Willnow TE. ApoE4 disrupts interaction of sortilin with fatty acid-binding protein 7 essential to promote lipid signaling. J Cell Sci 2021; 134:272562. [PMID: 34557909 PMCID: PMC8572006 DOI: 10.1242/jcs.258894] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 09/16/2021] [Indexed: 11/20/2022] Open
Abstract
Sortilin is a neuronal receptor for apolipoprotein E (apoE). Sortilin-dependent uptake of lipidated apoE promotes conversion of polyunsaturated fatty acids (PUFA) into neuromodulators that induce anti-inflammatory gene expression in the brain. This neuroprotective pathway works with the apoE3 variant but is lost with the apoE4 variant, the main risk factor for Alzheimer's disease (AD). Here, we elucidated steps in cellular handling of lipids through sortilin, and why they are disrupted by apoE4. Combining unbiased proteome screens with analyses in mouse models, we uncover interaction of sortilin with fatty acid-binding protein 7 (FABP7), the intracellular carrier for PUFA in the brain. In the presence of apoE3, sortilin promotes functional expression of FABP7 and its ability to elicit lipid-dependent gene transcription. By contrast, apoE4 binding blocks sortilin-mediated sorting, causing catabolism of FABP7 and impairing lipid signaling. Reduced FABP7 levels in the brain of AD patients expressing apoE4 substantiate the relevance of these interactions for neuronal lipid homeostasis. Taken together, we document interaction of sortilin with mediators of extracellular and intracellular lipid transport that provides a mechanistic explanation for loss of a neuroprotective lipid metabolism in AD.
Collapse
Affiliation(s)
- Antonino Asaro
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Rishabhdev Sinha
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Magda Bakun
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | | | | | - Tymon Rubel
- Warsaw University of Technology, Institute of Radioelectronics and Multimedia Technology, 00-665 Warsaw, Poland
| | - Annemieke Rozeboom
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, 1105AZ Amsterdam, The Netherlands.,Center for Neuroscience, Amsterdam Institute for Life Sciences, University of Amsterdam, 1098XH Amsterdam, The Netherlands
| | - Michal Dadlez
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland.,Biology Department, Institute of Genetics and Biotechnology02-106 Warsaw, Poland
| | - Bozena Kaminska
- Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Eleonora Aronica
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, 1105AZ Amsterdam, The Netherlands
| | - Anna R Malik
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany.,Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany.,Department of Medical Biochemistry, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
23
|
Larsen NY, Li X, Tan X, Ji G, Lin J, Rajkowska G, Møller J, Vihrs N, Sporring J, Sun F, Nyengaard JR. Cellular 3D-reconstruction and analysis in the human cerebral cortex using automatic serial sections. Commun Biol 2021; 4:1030. [PMID: 34475516 PMCID: PMC8413324 DOI: 10.1038/s42003-021-02548-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 08/09/2021] [Indexed: 12/17/2022] Open
Abstract
Techniques involving three-dimensional (3D) tissue structure reconstruction and analysis provide a better understanding of changes in molecules and function. We have developed AutoCUTS-LM, an automated system that allows the latest advances in 3D tissue reconstruction and cellular analysis developments using light microscopy on various tissues, including archived tissue. The workflow in this paper involved advanced tissue sampling methods of the human cerebral cortex, an automated serial section collection system, digital tissue library, cell detection using convolution neural network, 3D cell reconstruction, and advanced analysis. Our results demonstrated the detailed structure of pyramidal cells (number, volume, diameter, sphericity and orientation) and their 3D spatial organization are arranged in a columnar structure. The pipeline of these combined techniques provides a detailed analysis of tissues and cells in biology and pathology. Nick Larsen et al. developed a pipeline to collect and image serial sections from fixed human cortex, then apply deep learning to detect pyramidal cells from 3D reconstructions of these sections. Their results reiterate that cortical pyramidal cells are organized in a columnar structure and highlight the potential of this method, which is universally applicable to characterize cells for various tissues.
Collapse
Affiliation(s)
- Nick Y Larsen
- Core Centre for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark. .,Centre for Stochastic Geometry and Advanced Bioimaging, Aalborg University, Aarhus University and University of Copenhagen, Aalborg, Aarhus and Copenhagen, Denmark. .,Sino-Danish Center for Education and Research, Aarhus, Denmark. .,University of the Chinese Academy of Sciences, Beijing, China.
| | - Xixia Li
- National Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xueke Tan
- National Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Gang Ji
- National Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jing Lin
- Department of Computer Science, University of Copenhagen, Copenhagen, Denmark
| | - Grazyna Rajkowska
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jesper Møller
- Centre for Stochastic Geometry and Advanced Bioimaging, Aalborg University, Aarhus University and University of Copenhagen, Aalborg, Aarhus and Copenhagen, Denmark.,Department of Mathematical Sciences, Aalborg University, Aalborg, Denmark
| | - Ninna Vihrs
- Department of Mathematical Sciences, Aalborg University, Aalborg, Denmark
| | - Jon Sporring
- Centre for Stochastic Geometry and Advanced Bioimaging, Aalborg University, Aarhus University and University of Copenhagen, Aalborg, Aarhus and Copenhagen, Denmark.,Department of Computer Science, University of Copenhagen, Copenhagen, Denmark
| | - Fei Sun
- Sino-Danish Center for Education and Research, Aarhus, Denmark.,University of the Chinese Academy of Sciences, Beijing, China.,National Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jens R Nyengaard
- Core Centre for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Centre for Stochastic Geometry and Advanced Bioimaging, Aalborg University, Aarhus University and University of Copenhagen, Aalborg, Aarhus and Copenhagen, Denmark.,Sino-Danish Center for Education and Research, Aarhus, Denmark.,Department of Pathology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
24
|
In vivo functions of p75 NTR: challenges and opportunities for an emerging therapeutic target. Trends Pharmacol Sci 2021; 42:772-788. [PMID: 34334250 DOI: 10.1016/j.tips.2021.06.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/31/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022]
Abstract
The p75 neurotrophin receptor (p75NTR) functions at the molecular nexus of cell death, survival, and differentiation. In addition to its contribution to neurodegenerative diseases and nervous system injuries, recent studies have revealed unanticipated roles of p75NTR in liver repair, fibrinolysis, lung fibrosis, muscle regeneration, and metabolism. Linking these various p75NTR functions more precisely to specific mechanisms marks p75NTR as an emerging candidate for therapeutic intervention in a wide range of disorders. Indeed, small molecule inhibitors of p75NTR binding to neurotrophins have shown efficacy in models of Alzheimer's disease (AD) and neurodegeneration. Here, we outline recent advances in understanding p75NTR pleiotropic functions in vivo, and propose an integrated view of p75NTR and its challenges and opportunities as a pharmacological target.
Collapse
|
25
|
Covaceuszach S, Peche L, Konarev P, Lamba D. A combined evolutionary and structural approach to disclose the primary structural determinants essential for proneurotrophins biological functions. Comput Struct Biotechnol J 2021; 19:2891-2904. [PMID: 34094000 PMCID: PMC8144349 DOI: 10.1016/j.csbj.2021.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/03/2021] [Accepted: 05/03/2021] [Indexed: 12/24/2022] Open
Abstract
The neurotrophins, i.e., Nerve Growth Factor (NGF), Brain-Derived Neurotrophic Factor (BDNF), Neurotrophin 3 (NT3) and Neurotrophin 4 (NT4), are known to play a range of crucial functions in the developing and adult peripheral and central nervous systems. Initially synthesized as precursors, i.e., proneurotrophins (proNTs), that are cleaved to release C-terminal mature forms, they act through two types of receptors, the specific Trk receptors (Tropomyosin-related kinases) and the pan-neurotrophin receptor p75NTR, to initiate survival and differentiative responses. Recently, all the proNTs but proNT4 have been demonstrated to be not just inactive precursors, but signaling ligands that mediate opposing actions in fundamental aspects of the nervous system with respect to the mature counterparts through dual-receptor complexes formation with a member of the VPS10 family and p75NTR. Despite the functional relevance, the molecular determinants underpinning the interactions between the pro-domains and their receptors are still elusive probably due to their intrinsically disordered nature. Here we present an evolutionary approach coupled to an experimental study aiming to uncover the structural and dynamical basis of the biological function displayed by proNGF, proBDNF and proNT3 but missing in proNT4. A bioinformatic analysis allowed to elucidate the functional adaptability of the proNTs family in vertebrates, identifying conserved key structural features. The combined biochemical and SAXS experiments shed lights on the structure and dynamic behavior of the human proNTs in solution, giving insights on the evolutionary conserved structural motifs, essential for the multifaceted roles of proNTs in physiological as well as in pathological contexts.
Collapse
Affiliation(s)
- S. Covaceuszach
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Trieste, Italy
| | - L.Y. Peche
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Trieste, Italy
| | - P.V. Konarev
- A.V. Shubnikov Institute of Crystallography of Federal Scientific Research Centre “Crystallography and Photonics” of Russian Academy of Sciences, Moscow, Russia
| | - D. Lamba
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Trieste, Italy
- Interuniversity Consortium “Biostructures and Biosystems National Institute”, Roma, Italy
| |
Collapse
|
26
|
Ozalp O, Cark O, Azbazdar Y, Haykir B, Cucun G, Kucukaylak I, Alkan-Yesilyurt G, Sezgin E, Ozhan G. Nradd Acts as a Negative Feedback Regulator of Wnt/β-Catenin Signaling and Promotes Apoptosis. Biomolecules 2021; 11:100. [PMID: 33466728 PMCID: PMC7828832 DOI: 10.3390/biom11010100] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
Wnt/β-catenin signaling controls many biological processes for the generation and sustainability of proper tissue size, organization and function during development and homeostasis. Consequently, mutations in the Wnt pathway components and modulators cause diseases, including genetic disorders and cancers. Targeted treatment of pathway-associated diseases entails detailed understanding of the regulatory mechanisms that fine-tune Wnt signaling. Here, we identify the neurotrophin receptor-associated death domain (Nradd), a homolog of p75 neurotrophin receptor (p75NTR), as a negative regulator of Wnt/β-catenin signaling in zebrafish embryos and in mammalian cells. Nradd significantly suppresses Wnt8-mediated patterning of the mesoderm and neuroectoderm during zebrafish gastrulation. Nradd is localized at the plasma membrane, physically interacts with the Wnt receptor complex and enhances apoptosis in cooperation with Wnt/β-catenin signaling. Our functional analyses indicate that the N-glycosylated N-terminus and the death domain-containing C-terminus regions are necessary for both the inhibition of Wnt signaling and apoptosis. Finally, Nradd can induce apoptosis in mammalian cells. Thus, Nradd regulates cell death as a modifier of Wnt/β-catenin signaling during development.
Collapse
Affiliation(s)
- Ozgun Ozalp
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340 Izmir, Turkey; (O.O.); (O.C.); (Y.A.); (B.H.); (G.C.); (I.K.); (G.A.-Y.)
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340 Izmir, Turkey
| | - Ozge Cark
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340 Izmir, Turkey; (O.O.); (O.C.); (Y.A.); (B.H.); (G.C.); (I.K.); (G.A.-Y.)
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340 Izmir, Turkey
| | - Yagmur Azbazdar
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340 Izmir, Turkey; (O.O.); (O.C.); (Y.A.); (B.H.); (G.C.); (I.K.); (G.A.-Y.)
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340 Izmir, Turkey
| | - Betul Haykir
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340 Izmir, Turkey; (O.O.); (O.C.); (Y.A.); (B.H.); (G.C.); (I.K.); (G.A.-Y.)
- Institute of Physiology, Switzerland and National Center of Competence in Research NCCR Kidney, University of Zurich, CH-8057 Zurich, Switzerland
| | - Gokhan Cucun
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340 Izmir, Turkey; (O.O.); (O.C.); (Y.A.); (B.H.); (G.C.); (I.K.); (G.A.-Y.)
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340 Izmir, Turkey
| | - Ismail Kucukaylak
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340 Izmir, Turkey; (O.O.); (O.C.); (Y.A.); (B.H.); (G.C.); (I.K.); (G.A.-Y.)
- Institute of Zoology-Developmental Biology, University of Cologne, 50674 Cologne, Germany
| | - Gozde Alkan-Yesilyurt
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340 Izmir, Turkey; (O.O.); (O.C.); (Y.A.); (B.H.); (G.C.); (I.K.); (G.A.-Y.)
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden;
- MRC Weatherall Institute of Molecular Medicine, MRC Human Immunology Unit, University of Oxford, Oxford OX39DS, UK
| | - Gunes Ozhan
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340 Izmir, Turkey; (O.O.); (O.C.); (Y.A.); (B.H.); (G.C.); (I.K.); (G.A.-Y.)
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340 Izmir, Turkey
| |
Collapse
|
27
|
Zhou X, Kukar T, Rademakers R. Lysosomal Dysfunction and Other Pathomechanisms in FTLD: Evidence from Progranulin Genetics and Biology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:219-242. [PMID: 33433878 DOI: 10.1007/978-3-030-51140-1_14] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
It has been more than a decade since heterozygous loss-of-function mutations in the progranulin gene (GRN) were first identified as an important genetic cause of frontotemporal lobar degeneration (FTLD). Due to the highly diverse biological functions of the progranulin (PGRN) protein, encoded by GRN, multiple possible disease mechanisms have been proposed. Early work focused on the neurotrophic properties of PGRN and its role in the inflammatory response. However, since the discovery of homozygous GRN mutations in patients with a lysosomal storage disorder, investigation into the possible roles of PGRN and its proteolytic cleavage products granulins, in lysosomal function and dysfunction, has taken center stage. In this chapter, we summarize the GRN mutational spectrum and its associated phenotypes followed by an in-depth discussion on the possible disease mechanisms implicated in FTLD-GRN. We conclude with key outstanding questions which urgently require answers to ensure safe and successful therapy development for GRN mutation carriers.
Collapse
Affiliation(s)
- Xiaolai Zhou
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Thomas Kukar
- Department of Pharmacology and Chemical Biology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- VIB Center for Molecular Neurology, University of Antwerp-CDE, Antwerp, Belgium.
| |
Collapse
|
28
|
Intervention of Brain-Derived Neurotrophic Factor and Other Neurotrophins in Adult Neurogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1331:95-115. [PMID: 34453295 DOI: 10.1007/978-3-030-74046-7_8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell survival during adult neurogenesis and the modulation of each step, namely, proliferation, lineage differentiation, migration, maturation, and functional integration of the newborn cells into the existing circuitry, is regulated by intrinsic and extrinsic factors. Transduction of extracellular niche signals triggers the activation of intracellular mechanisms that regulate adult neurogenesis by affecting gene expression. While the intrinsic factors include transcription factors and epigenetic regulators, the extrinsic factors are molecular signals that are present in the neurogenic niche microenvironment. These include morphogens, growth factors, neurotransmitters, and signaling molecules secreted as soluble factors or associated to the extracellular matrix. Among these molecular mechanisms are neurotrophins and neurotrophin receptors which have been implicated in the regulation of adult neurogenesis at different levels, with brain-derived neurotrophic factor (BDNF) being the most studied neurotrophin. In this chapter, we review the current knowledge about the role of neurotrophins in the regulation of adult neurogenesis in both the subventricular zone (SVZ) and the hippocampal subgranular zone (SGZ).
Collapse
|
29
|
c-Jun N-terminal Kinase Mediates Ligand-independent p75 NTR Signaling in Mesencephalic Cells Subjected to Oxidative Stress. Neuroscience 2020; 453:222-236. [PMID: 33253821 DOI: 10.1016/j.neuroscience.2020.11.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 11/01/2020] [Accepted: 11/22/2020] [Indexed: 12/11/2022]
Abstract
The p75 neurotrophin receptor (p75NTR) is a multifunctional protein that regulates cellular responses to pathological conditions in specific regions of the nervous system. Activation of p75NTR in certain neuronal populations induces proteolytic processing of the receptor, thereby generating p75NTR fragments that facilitate downstream signaling. Expression of p75NTR has been reported in neurons of the ventral midbrain, but p75NTR signaling mechanisms in such cells are poorly understood. Here, we used Lund Human Mesencephalic cells, a population of neuronal cells derived from the ventral mesencephalon, to evaluate the effects of oxidative stress on p75NTR signaling. Subjection of the cells to oxidative stress resulted in decreased cell-surface localization of p75NTR and intracellular accumulation of p75NTR fragments. Oxidative stress-induced p75NTR processing was reduced by pharmacological inhibition of metalloproteases or γ-secretase, but was unaltered by blockade of the ligand-binding domain of p75NTR. Furthermore, inhibition of c-Jun N-terminal Kinase (JNK) decreased p75NTR cleavage induced by oxidative damage. Altogether, these results support a mechanism of p75NTR activation in which oxidative stress stimulates JNK signaling, thereby facilitating p75NTR processing via a ligand-independent mechanism involving induction of metalloprotease and γ-secretase activity. These findings reveal a novel role for JNK in ligand-independent p75NTR signaling, and, considering the susceptibility of mesencephalic neurons to oxidative damage associated with Parkinson's disease (PD), merit further investigation into the effects of p75NTR on PD-related neurodegeneration.
Collapse
|
30
|
VPS10P Domain Receptors: Sorting Out Brain Health and Disease. Trends Neurosci 2020; 43:870-885. [DOI: 10.1016/j.tins.2020.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/23/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022]
|
31
|
Saiyasit N, Chunchai T, Apaijai N, Pratchayasakul W, Sripetchwandee J, Chattipakorn N, Chattipakorn SC. Chronic high-fat diet consumption induces an alteration in plasma/brain neurotensin signaling, metabolic disturbance, systemic inflammation/oxidative stress, brain apoptosis, and dendritic spine loss. Neuropeptides 2020; 82:102047. [PMID: 32327191 DOI: 10.1016/j.npep.2020.102047] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/11/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023]
Abstract
Chronic high-fat diet (HFD) consumption caused not only negative effects on obesity and metabolic disturbance, but also instigated several brain pathologies, including dendritic spine loss. In addition, alterations in plasma/brain neurotensin (NT) levels and NT signaling were observed in obesity. However, the mechanistic link between the NT levels in plasma and brain, NT signaling, and peripheral/brain pathologies following prolonged HFD consumption still needs to be elucidated. We hypothesized that an increase in peripheral/brain NT signaling were associated with peripheral/brain pathologies after prolonged HFD consumption. Male Wistar rats (n = 24) were given either a normal diet (ND) or a HFD for 12 and 40 weeks. At the end of each time course, metabolic parameters and plasma NT levels were measured. Rats were then decapitated and the brains were examined the levels of brain NT, hippocampal reactive oxygen species, the number of Iba-1 positive cells, the dendritic spine densities, and the expression of NT-, mitophagy-, autophagy-, and apoptotic-related proteins. The findings showed an increase in the level of plasma NT with dyslipidemia, metabolic disturbances, systemic inflammation/oxidative stress, and hippocampal pathologies in rats fed HFD for 12 and 40 weeks. The expression of brain NT signaling and brain apoptosis were markedly increased after 40 weeks of HFD feeding. These results indicated that the alteration in the level of circulating/brain NT and its downstream signaling were associated with central and peripheral pathologies after long-term HFD intake. Therefore, these alterations in NT level or its signaling could be considered as a therapeutic target in treating obesity.
Collapse
Affiliation(s)
- Napatsorn Saiyasit
- Neurophysiology Unit, Cardiac Electrophysiology Research, and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Titikorn Chunchai
- Neurophysiology Unit, Cardiac Electrophysiology Research, and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research, and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wasana Pratchayasakul
- Neurophysiology Unit, Cardiac Electrophysiology Research, and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jirapas Sripetchwandee
- Neurophysiology Unit, Cardiac Electrophysiology Research, and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research, and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research, and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
32
|
Monti G, Jensen ML, Mehmedbasic A, Jørgensen MM, Holm IE, Barkholt P, Zole E, Vægter CB, Vorum H, Nyengaard JR, Andersen OM. SORLA Expression in Synaptic Plexiform Layers of Mouse Retina. Mol Neurobiol 2020; 57:3106-3117. [PMID: 32472518 DOI: 10.1007/s12035-020-01946-x] [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: 12/18/2019] [Accepted: 05/13/2020] [Indexed: 11/25/2022]
Abstract
Sorting protein-related receptor containing LDLR class A repeats (SORLA; also known as LR11) exerts intraneuronal trafficking functions in the central nervous system. Recently, involvement of SORLA in retinogenesis was proposed, but no studies have examined yet in detail the expression pattern of this sorting receptor in the retina. Here, we provide a spatio-temporal characterization of SORL1 mRNA and its translational product SORLA in the postnatal mouse retina. Using stereological analysis, we confirmed previous studies showing that receptor depletion in knockout mice significantly reduces the number of cells in the inner nuclear layer (INL), suggesting that functional SORLA expression is essential for the development of this retinal strata. qPCR and Western blot analyses showed that SORL1/SORLA expression peaks at postnatal day 15, just after eye opening. Interestingly, we found that transcripts are somatically located in several neuronal populations residing in the INL and the ganglion cell layer, whereas SORLA protein is also present in the synaptic plexiform layers. In line with receptor expression in dendritic terminals, we found delayed stratification of the inner plexiform layer in knockout mice, indicating an involvement of SORLA in neuronal connectivity. Altogether, these data suggest a novel role of SORLA in synaptogenesis. Receptor dysfunctions may be implicated in morphological and functional impairments of retinal inner layer formation associated with eye disorders.
Collapse
Affiliation(s)
- Giulia Monti
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Marianne L Jensen
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Arnela Mehmedbasic
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Margarita Melnikova Jørgensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Pathology, Randers Regional Hospital, Randers, Denmark
| | - Ida E Holm
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Pathology, Randers Regional Hospital, Randers, Denmark
| | - Pernille Barkholt
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Egija Zole
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Christian B Vægter
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Henrik Vorum
- Department of Ophthalmology, Aalborg University Hospital, Hobrovej 18-22, DK-9000, Aalborg, Denmark
| | - Jens R Nyengaard
- Core Centre for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Olav M Andersen
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark.
| |
Collapse
|
33
|
MicroRNA-148a regulates low-density lipoprotein metabolism by repressing the (pro)renin receptor. PLoS One 2020; 15:e0225356. [PMID: 32437440 PMCID: PMC7241754 DOI: 10.1371/journal.pone.0225356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 05/06/2020] [Indexed: 01/15/2023] Open
Abstract
High plasma LDL cholesterol (LDL-c) concentration is a major risk factor for atherosclerosis. Hepatic LDL receptor (LDLR) regulates LDL metabolism, and thereby plasma LDL-c concentration. Recently, we have identified the (pro)renin receptor [(P)RR] as a novel regulator of LDL metabolism, which regulates LDLR degradation and hence its protein abundance and activity. In silico analysis suggests that the (P)RR is a target of miR-148a. In this study we determined whether miR-148a could regulate LDL metabolism by regulating (P)RR expression in HepG2 and Huh7 cells. We found that miR-148a suppressed (P)RR expression by binding to the 3’-untranslated regions (3’-UTR) of the (P)RR mRNA. Mutating the binding sites for miR-148a in the 3’-UTR of (P)RR mRNA completely abolished the inhibitory effects of miR-148a on (P)RR expression. In line with our recent findings, reduced (P)RR expression resulted in decreased cellular LDL uptake, likely as a consequence of decreased LDLR protein abundance. Overexpressing the (P)RR prevented miR-148a-induced reduction in LDLR abundance and cellular LDL uptake. Our study supports a new concept that miR-148a is a regulator of (P)RR expression. By reducing (P)RR abundance, miR-148a decreases LDLR protein abundance and consequently cellular LDL uptake.
Collapse
|
34
|
Kuhn KD, Edamura K, Bhatia N, Cheng I, Clark SA, Haynes CV, Heffner DL, Kabir F, Velasquez J, Spano AJ, Deppmann CD, Keeler AB. Molecular dissection of TNFR-TNFα bidirectional signaling reveals both cooperative and antagonistic interactions with p75 neurotrophic factor receptor in axon patterning. Mol Cell Neurosci 2020; 103:103467. [PMID: 32004684 PMCID: PMC7682658 DOI: 10.1016/j.mcn.2020.103467] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/18/2019] [Accepted: 01/13/2020] [Indexed: 11/25/2022] Open
Abstract
During neural development, complex organisms rely on progressive and regressive events whereby axons, synapses, and neurons are overproduced followed by selective elimination of a portion of these components. Tumor necrosis factor α (TNFα) together with its cognate receptor (Tumor necrosis factor receptor 1; TNFR1) have been shown to play both regressive (i.e. forward signaling from the receptor) and progressive (i.e. reverse signaling from the ligand) roles in sympathetic neuron development. In contrast, a paralog of TNFR1, p75 neurotrophic factor receptor (p75NTR) promotes mainly regressive developmental events in sympathetic neurons. Here we examine the interplay between these paralogous receptors in the regulation of axon branch elimination and arborization. We confirm previous reports that these TNFR1 family members are individually capable of promoting ligand-dependent suppression of axon growth and branching. Remarkably, p75NTR and TNFR1 physically interact and p75NTR requires TNFR1 for ligand-dependent axon suppression of axon branching but not vice versa. We also find that p75NTR forward signaling and TNFα reverse signaling are functionally antagonistic. Finally, we find that TNFα reverse signaling is necessary for nerve growth factor (NGF) dependent axon growth. Taken together these findings demonstrate several levels of synergistic and antagonistic interactions using very few signaling pathways and that the balance of these synergizing and opposing signals act to ensure proper axon growth and patterning.
Collapse
Affiliation(s)
- K D Kuhn
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - K Edamura
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - N Bhatia
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - I Cheng
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - S A Clark
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - C V Haynes
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - D L Heffner
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - F Kabir
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - J Velasquez
- Blue Ridge Virtual Governor's School, Palmyra, VA 22963, USA
| | - A J Spano
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - C D Deppmann
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, USA; Department of Cell Biology, University of Virginia, Charlottesville, VA 22903, USA.
| | - A B Keeler
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA.
| |
Collapse
|
35
|
Saragovi HU, Galan A, Levin LA. Neuroprotection: Pro-survival and Anti-neurotoxic Mechanisms as Therapeutic Strategies in Neurodegeneration. Front Cell Neurosci 2019; 13:231. [PMID: 31244606 PMCID: PMC6563757 DOI: 10.3389/fncel.2019.00231] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/08/2019] [Indexed: 12/14/2022] Open
Abstract
Neurotrophins (NTs) are a subset of the neurotrophic factor family. These growth factors were originally named based on the nerve growth functional assays used to identify them. NTs act as paracrine or autocrine factors for cells expressing NT receptors. The receptors and their function have been studied primarily in cells of the nervous system, but are also present in the cardiovascular, endocrine, and immune systems, as well as in many neoplastic cells. The signals activated by NTs can be varied, depending on cellular stage and context, healthy or disease states, and depending on whether the specific NTs and their receptors are expressed in the relevant cells. In the healthy central and peripheral adult nervous systems, NTs drive neuronal survival, phenotype, synaptic maintenance, and function. Deficiencies of the NT/NT receptor axis are causally associated with disease onset or disease progression. Paradoxically, NTs can also drive synaptic loss and neuronal death. In the embryonic stage this activity is essential for proper developmental pruning of the nervous system, but in the adult it can be associated with neurodegenerative disease. Given their key role in neuronal survival and death, NTs and NT receptors have long been considered therapeutic targets to achieve neuroprotection. The first neuroprotective approaches consisted of enhancing neuronal survival signals using NTs. Later strategies selectively targeted receptors to induce survival signals specifically, while avoiding activation of death signals. Recently, the concept of selectively targeting receptors to reduce neuronal death signals has emerged. Here, we review the rationale of each neuroprotective strategy with respect to the complex cell biology and pharmacology of each target receptor.
Collapse
Affiliation(s)
- Horacio Uri Saragovi
- Lady Davis Institute, Montreal, QC, Canada.,Jewish General Hospital, Montreal, QC, Canada.,Department of Ophthalmology and Visual Sciences, McGill University, Montreal, QC, Canada
| | - Alba Galan
- Lady Davis Institute, Montreal, QC, Canada.,Jewish General Hospital, Montreal, QC, Canada
| | - Leonard A Levin
- Department of Ophthalmology and Visual Sciences, McGill University, Montreal, QC, Canada.,McGill University Health Centre, Montreal, QC, Canada.,Montreal Neurological Institute, Mcgill University, Montreal, QC, Canada
| |
Collapse
|
36
|
Svobodova B, Kloudova A, Ruzicka J, Kajtmanova L, Navratil L, Sedlacek R, Suchy T, Jhanwar-Uniyal M, Jendelova P, Machova Urdzikova L. The effect of 808 nm and 905 nm wavelength light on recovery after spinal cord injury. Sci Rep 2019; 9:7660. [PMID: 31113985 PMCID: PMC6529518 DOI: 10.1038/s41598-019-44141-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 05/08/2019] [Indexed: 01/01/2023] Open
Abstract
We investigated the effect of a Multiwave Locked System laser (with a simultaneous 808 nm continuous emission and 905 nm pulse emission) on the spinal cord after spinal cord injury (SCI) in rats. The functional recovery was measured by locomotor tests (BBB, Beam walking, MotoRater) and a sensitivity test (Plantar test). The locomotor tests showed a significant improvement of the locomotor functions of the rats after laser treatment from the first week following lesioning, compared to the controls. The laser treatment significantly diminished thermal hyperalgesia after SCI as measured by the Plantar test. The atrophy of the soleus muscle was reduced in the laser treated rats. The histopathological investigation showed a positive effect of the laser therapy on white and gray matter sparing. Our data suggests an upregulation of M2 macrophages in laser treated animals by the increasing number of double labeled CD68+/CD206+ cells in the cranial and central parts of the lesion, compared to the control animals. A shift in microglial/macrophage polarization was confirmed by gene expression analysis by significant mRNA downregulation of Cd86 (marker of inflammatory M1), and non-significant upregulation of Arg1 (marker of M2). These results demonstrated that the combination of 808 nm and 905 nm wavelength light is a promising non-invasive therapy for improving functional recovery and tissue sparing after SCI.
Collapse
Affiliation(s)
- Barbora Svobodova
- Institute of Experimental Medicine, Academy of Sciences, Prague, Czech Republic.,2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Anna Kloudova
- Institute of Experimental Medicine, Academy of Sciences, Prague, Czech Republic
| | - Jiri Ruzicka
- Institute of Experimental Medicine, Academy of Sciences, Prague, Czech Republic
| | | | - Leos Navratil
- Department of Health Care Disciplines and Population Protection, Faculty of Biomedical Engineering, Czech Technical University, Kladno, Czech Republic
| | - Radek Sedlacek
- Laboratory of Biomechanics, Department of Mechanics, Biomechanics and Mechatronics, Faculty of Mechanical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Tomas Suchy
- Laboratory of Biomechanics, Department of Mechanics, Biomechanics and Mechatronics, Faculty of Mechanical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | | | - Pavla Jendelova
- Institute of Experimental Medicine, Academy of Sciences, Prague, Czech Republic. .,2nd Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Lucia Machova Urdzikova
- Institute of Experimental Medicine, Academy of Sciences, Prague, Czech Republic. .,2nd Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
37
|
Investigating the Conformational Response of the Sortilin Receptor upon Binding Endogenous Peptide- and Protein Ligands by HDX-MS. Structure 2019; 27:1103-1113.e3. [PMID: 31104815 DOI: 10.1016/j.str.2019.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/28/2019] [Accepted: 04/10/2019] [Indexed: 11/20/2022]
Abstract
Sortilin is a multifunctional neuronal receptor involved in sorting of neurotrophic factors and apoptosis signaling. So far, structural characterization of sortilin and its endogenous ligands has been limited to crystallographic studies of sortilin in complex with the neuropeptide neurotensin. Here, we use hydrogen/deuterium exchange mass spectrometry to investigate the conformational response of sortilin to binding biological ligands including the peptides neurotensin and the sortilin propeptide and the proteins progranulin and pro-nerve growth factor-β. The results show that the ligands use two binding sites inside the cavity of the β-propeller of sortilin. However, ligands have distinct differences in their conformational impact on the receptor. Interestingly, the protein ligands induce conformational stabilization in a remote membrane-proximal domain, hinting at an unknown conformational link between the ligand binding region and this membrane-proximal region of sortilin. Our findings improve our structural understanding of sortilin and how it mediates diverse ligand-dependent functions important in neurobiology.
Collapse
|
38
|
Wu LS, Cheng WC, Chen CY, Wu MC, Wang YC, Tseng YH, Chuang TJ, Shen CKJ. Transcriptomopathies of pre- and post-symptomatic frontotemporal dementia-like mice with TDP-43 depletion in forebrain neurons. Acta Neuropathol Commun 2019; 7:50. [PMID: 30922385 PMCID: PMC6440020 DOI: 10.1186/s40478-019-0674-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 02/04/2019] [Indexed: 12/12/2022] Open
Abstract
TAR DNA-binding protein (TDP-43) is a ubiquitously expressed nuclear protein, which participates in a number of cellular processes and has been identified as the major pathological factor in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Here we constructed a conditional TDP-43 mouse with depletion of TDP-43 in the mouse forebrain and find that the mice exhibit a whole spectrum of age-dependent frontotemporal dementia-like behaviour abnormalities including perturbation of social behaviour, development of dementia-like behaviour, changes of activities of daily living, and memory loss at a later stage of life. These variations are accompanied with inflammation, neurodegeneration, and abnormal synaptic plasticity of the mouse CA1 neurons. Importantly, analysis of the cortical RNA transcripts of the conditional knockout mice at the pre-/post-symptomatic stages and the corresponding wild type mice reveals age-dependent alterations in the expression levels and RNA processing patterns of a set of genes closely associated with inflammation, social behaviour, synaptic plasticity, and neuron survival. This study not only supports the scenario that loss-of-function of TDP-43 in mice may recapitulate key behaviour features of the FTLD diseases, but also provides a list of TDP-43 target genes/transcript isoforms useful for future therapeutic research.
Collapse
Affiliation(s)
- Lien-Szu Wu
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, 115, Taiwan, Republic of China
| | - Wei-Cheng Cheng
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, 115, Taiwan, Republic of China
| | - Chia-Ying Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Ming-Che Wu
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, 115, Taiwan, Republic of China
| | - Yi-Chi Wang
- Research Center for Environmental Changes, Academia Sinica, Taipei, Taiwan, Republic of China
| | | | | | - C-K James Shen
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, 115, Taiwan, Republic of China.
| |
Collapse
|
39
|
Segatto M, Fico E, Gharbiya M, Rosso P, Carito V, Tirassa P, Plateroti R, Lambiase A. VEGF inhibition alters neurotrophin signalling pathways and induces caspase-3 activation and autophagy in rabbit retina. J Cell Physiol 2019; 234:18297-18307. [PMID: 30891770 DOI: 10.1002/jcp.28462] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/26/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022]
Abstract
This study sought to evaluate the prospective role exerted by vascular endothelial growth factor (VEGF) in the modulation of nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) signalling pathways in the rabbit retina. To reach this aim, the anti-VEGF agents aflibercept and ranibizumab were used as a pharmacological approach to evaluate the putative consequences elicited by VEGF inhibition on neurotrophin signalling. VEGF inhibition determined a marked imbalance in proneurotrophin expression, a significant reduction in TrkA and TrkB phosphorylation states and a decrease in the pan-neurotrophin receptor p75. Importantly, VEGF blockade also caused a strong increase in cleaved caspase-3, beclin-1 and lipidated LC3. The effects were more pronounced in the aflibercept group when compared with ranibizumab-treated rabbits, particularly 1 week after injection. This study demonstrates that VEGF exerts pivotal physiological roles in regulating NGF and BDNF pathways in the retina, as its inhibition by anti-VEGF agents deeply impacts neurotrophin homeostasis. These events are accompanied by a sustained induction of apoptotic and autophagic markers, suggesting that anti-VEGF-dependent impairments in neurotrophin signalling could be responsible for the activation of retinal cell death pathways.
Collapse
Affiliation(s)
- Marco Segatto
- Department of Biosciences and Territory, University of Molise, Pesche, Italy.,Department of Sense Organs, University of Rome "La Sapienza", Rome, Italy
| | - Elena Fico
- Institute of Cell Biology and Neurobiology (IBCN-CNR), Rome, Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Magda Gharbiya
- Department of Sense Organs, University of Rome "La Sapienza", Rome, Italy
| | - Pamela Rosso
- Institute of Cell Biology and Neurobiology (IBCN-CNR), Rome, Italy
| | - Valentina Carito
- Institute of Cell Biology and Neurobiology (IBCN-CNR), Rome, Italy
| | - Paola Tirassa
- Institute of Cell Biology and Neurobiology (IBCN-CNR), Rome, Italy
| | - Rocco Plateroti
- Department of Sense Organs, University of Rome "La Sapienza", Rome, Italy
| | | |
Collapse
|
40
|
Yang W, Wu PF, Ma JX, Liao MJ, Wang XH, Xu LS, Xu MH, Yi L. Sortilin promotes glioblastoma invasion and mesenchymal transition through GSK-3β/β-catenin/twist pathway. Cell Death Dis 2019; 10:208. [PMID: 30814514 PMCID: PMC6393543 DOI: 10.1038/s41419-019-1449-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 02/02/2019] [Accepted: 02/12/2019] [Indexed: 12/21/2022]
Abstract
High aggressiveness is a hallmark of glioblastoma and predicts poor prognosis of patients with glioblastoma. The expression level of sortilin has been preliminarily reported to be elevated in high-grade glioma; however, the potential significance of sortilin in glioblastoma progression has not been elucidated. In this study, we investigated the oncogenic effect of sortilin in glioblastoma. Increased levels of sortilin were noted in the mesenchymal subtype of glioblastoma and highly aggressive subtypes of glioblastoma tissues and cell lines. In addition, high levels of sortilin predicted poor prognoses in patients with glioblastoma. Sortilin knockdown or inhibition with AF38469 (an orally bioavailable inhibitor of sortilin) significantly suppressed migration and invasion by inhibiting EMT-like mesenchymal transition in glioblastoma cells. Furthermore, we proved that sortilin promoted cell invasion mainly via Glycogen synthase kinase 3 beta (GSK-3β)/β-catenin/Twist-induced EMT-like mesenchymal transition in glioblastoma. Taken together, our results demonstrate a critical role of sortilin in glioblastoma invasion and EMT-like mesenchymal transition, indicating that sortilin contributes to glioblastoma progression. These data also highlight the dramatic antitumor effects of AF38469 in glioblastoma, suggesting that AF38469 is a potentially powerful antitumor agent for sortilin-overexpressing human glioblastoma.
Collapse
Affiliation(s)
- Wei Yang
- Department of Neurosurgery, Daping Hospital and Institute Research of Surgery, Army Medical University, Chongqing, 400042, China
| | - Peng-Fei Wu
- Department of Neurosurgery, Daping Hospital and Institute Research of Surgery, Army Medical University, Chongqing, 400042, China
| | - Jian-Xing Ma
- Department of Neurosurgery, Daping Hospital and Institute Research of Surgery, Army Medical University, Chongqing, 400042, China
| | - Mao-Jun Liao
- Department of Neurosurgery, Daping Hospital and Institute Research of Surgery, Army Medical University, Chongqing, 400042, China
| | - Xu-Hui Wang
- Department of Neurosurgery, Daping Hospital and Institute Research of Surgery, Army Medical University, Chongqing, 400042, China
| | - Lun-Shan Xu
- Department of Neurosurgery, Daping Hospital and Institute Research of Surgery, Army Medical University, Chongqing, 400042, China
| | - Min-Hui Xu
- Department of Neurosurgery, Daping Hospital and Institute Research of Surgery, Army Medical University, Chongqing, 400042, China.
| | - Liang Yi
- Department of Neurosurgery, Daping Hospital and Institute Research of Surgery, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
41
|
Montroull LE, Danelon V, Cragnolini AB, Mascó DH. Loss of TrkB Signaling Due to Status Epilepticus Induces a proBDNF-Dependent Cell Death. Front Cell Neurosci 2019; 13:4. [PMID: 30800056 PMCID: PMC6375841 DOI: 10.3389/fncel.2019.00004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 01/08/2019] [Indexed: 01/25/2023] Open
Abstract
Neurotrophins (NTs) are secretory proteins that bind to target receptors and influence many cellular functions, such as cell survival and cell death in neurons. The mammalian NT brain-derived neurotrophic factor (matBDNF) is the C-terminal mature form released by cleavage from the proBDNF precursor. The binding of matBDNF to the tyrosine kinase receptor B (TrkB) activates different signaling cascades and leads to neuron survival and plasticity, while the interaction of proBDNF with the p75 NT receptor (p75NTR)/sortilin receptor complex has been highly involved in apoptosis. Many studies have demonstrated that prolonged seizures such as status epilepticus (SE) induce changes in the expression of NT, pro-NT, and their receptors. We have previously described that the blockage of both matBDNF and proBDNF signaling reduces neuronal death after SE in vivo (Unsain et al., 2008). We used an in vitro model as well as an in vivo model of SE to determine the specific role of TrkB and proBDNF signaling during neuronal cell death. We found that the matBDNF sequestering molecule TrkB-Fc induced an increase in neuronal death in both models of SE, and it also prevented a decrease in TrkB levels. Moreover, SE triggered the interaction between proBDNF and p75NTR, which was not altered by sequestering matBDNF. The intra-hippocampal administration of TrkB-Fc, combined with an antibody against proBDNF, prevented neuronal degeneration. In addition, we demonstrated that proBDNF binding to p75NTR exacerbates neuronal death when matBDNF signaling is impaired through TrkB. Our results indicated that both the mature and the precursor forms of BDNF may have opposite effects depending on the scenario in which they function and the signaling pathways they activate.
Collapse
Affiliation(s)
- Laura Ester Montroull
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba; Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Víctor Danelon
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba; Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Andrea Beatriz Cragnolini
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba; Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Daniel Hugo Mascó
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba; Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
42
|
Hasselholt S, Hahn U, Vedel Jensen EB, Nyengaard JR. Practical implementation of the planar and spatial rotator in a complex tissue: the brain. J Microsc 2018; 273:26-35. [PMID: 30240001 DOI: 10.1111/jmi.12757] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 11/29/2022]
Abstract
In neuroscience, application of widely used stereological local volume estimators, including the planar rotator, is challenged by the combination of a complex tissue organisation and an estimator requirement of either isotropic or vertical sections, i.e. randomly oriented tissue. The spatial rotator is applicable with any tissue orientation but is sensitive to projection artefacts. The challenge is thus to select the most appropriate method for individual analyses. In this study, agreement between estimates of mean cell volume acquired with the vertical planar and the spatial rotator is assessed for two brain regions with different types of cytoarchitecture (motor cortex and hippocampal cornu ammonis 1). The possibility of using the planar rotator in tissues cut in an arbitrary direction is explored and requirements for a theoretically unbiased result as well as histological considerations are provided. LAY DESCRIPTION: Cells may change volume both during disease and with advancing age. Assessment of the volume of individual cells can therefore serve as a useful indicator of general tissue state. Most available methods to estimate cell volume in tissue sections, however, require that the tissue analysed has random orientation. Particularly for complex tissues such as the brain this is a challenge as identification, delineation and subdivision of many brain areas rely heavily on the use of anatomical atlases where illustrations depict the tissue in a few well-known orientations. In this study, the practical application of two different methods for estimating mean cell volumes in tissues cut in a preferred orientation is evaluated. Requirements for the feasibility of cell volume estimation without random tissue orientation as well as histological considerations are provided.
Collapse
Affiliation(s)
- S Hasselholt
- Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark.,Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Sino-Danish Center for Education and Research (SDC), Aarhus, Denmark
| | - U Hahn
- Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark.,Department of Mathematics, Aarhus University, Aarhus, Denmark
| | - E B Vedel Jensen
- Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark.,Department of Mathematics, Aarhus University, Aarhus, Denmark
| | - J R Nyengaard
- Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark.,Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Sino-Danish Center for Education and Research (SDC), Aarhus, Denmark
| |
Collapse
|
43
|
Francis Stuart SD, Wang L, Woodard WR, Ng GA, Habecker BA, Ripplinger CM. Age-related changes in cardiac electrophysiology and calcium handling in response to sympathetic nerve stimulation. J Physiol 2018; 596:3977-3991. [PMID: 29938794 DOI: 10.1113/jp276396] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/22/2018] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS Ageing results in changes to cardiac electrophysiology, Ca2+ handling, and β-adrenergic responsiveness. Sympathetic neurodegeneration also occurs with age, yet detailed action potential and Ca2+ handling responses to physiological sympathetic nerve stimulation (SNS) in the aged heart have not been assessed. Optical mapping in mouse hearts with intact sympathetic innervation revealed reduced responsiveness to SNS in the aged atria (assessed by heart rate) and aged ventricles (assessed by action potentials and Ca2+ transients). Sympathetic nerve density and noradrenaline content were reduced in aged ventricles, but noradrenaline content was preserved in aged atria. These results demonstrate that reduced responsiveness to SNS in the atria may be primarily due to decreased β-adrenergic receptor responsiveness, whereas reduced responsiveness to SNS in the ventricles may be primarily due to neurodegeneration. ABSTRACT The objective of this study was to determine how age-related changes in sympathetic structure and function impact cardiac electrophysiology and intracellular Ca2+ handling. Innervated hearts from young (3-4 months, YWT, n = 10) and aged (20-24 months, AGED, n = 11) female mice (C57Bl6) were optically mapped using the voltage (Vm ,)- and calcium (Ca2+ )-sensitive indicators Rh237 and Rhod2-AM. Sympathetic nerve stimulation (SNS) was performed at the spinal cord (T1-T3). β-Adrenergic responsiveness was assessed with isoproterenol (1 μM, ISO). Sympathetic nerve density and noradrenaline content were also quantified. Stimulation thresholds necessary to produce a defined increase in heart rate (HR) with SNS were higher in AGED vs. YWT hearts (5.4 ± 0.4 vs. 3.8 ± 0.4 Hz, P < 0.05). Maximal HR with SNS was lower in AGED vs. YWT (20.5 ± 3.41% vs. 73.0 ± 7.63% increase, P < 0.05). β-Adrenergic responsiveness of the atria (measured as percentage increase in HR with ISO) was decreased in AGED vs. YWT hearts (75.3 ± 22.5% vs. 148.5 ± 19.8%, P < 0.05). SNS significantly increased action potential duration (APD) in YWT but not AGED. Ca2+ transient durations and rise times were unchanged by SNS, yet AGED hearts had an increased susceptibility to Ca2+ alternans and ventricular arrhythmias. β-Adrenergic responsiveness of all ventricular parameters were similar between AGED and YWT. Sympathetic nerve density and noradrenaline content were decreased in the AGED ventricle, but not atria, compared to YWT. These data suggest that decreased responsiveness to SNS in the aged atria may be primarily due to decreased β-adrenergic responsiveness, whereas decreased responsiveness to SNS in the aged ventricles may be primarily due to nerve degeneration.
Collapse
Affiliation(s)
| | - Lianguo Wang
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - William R Woodard
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR, USA
| | - G Andre Ng
- Department of Cardiovascular Sciences, University of Leicester, NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Beth A Habecker
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR, USA
| | | |
Collapse
|
44
|
NG2/CSPG4 and progranulin in the posttraumatic glial scar. Matrix Biol 2018; 68-69:571-588. [DOI: 10.1016/j.matbio.2017.10.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/05/2017] [Accepted: 10/06/2017] [Indexed: 12/17/2022]
|
45
|
Angiotensin generation in the brain: a re-evaluation. Clin Sci (Lond) 2018; 132:839-850. [PMID: 29712882 DOI: 10.1042/cs20180236] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 02/06/2023]
Abstract
The existence of a so-called brain renin-angiotensin system (RAS) is controversial. Given the presence of the blood-brain barrier, angiotensin generation in the brain, if occurring, should depend on local synthesis of renin and angiotensinogen. Yet, although initially brain-selective expression of intracellular renin was reported, data in intracellular renin knockout animals argue against a role for this renin in angiotensin generation. Moreover, renin levels in brain tissue at most represented renin in trapped blood. Additionally, in neurogenic hypertension brain prorenin up-regulation has been claimed, which would generate angiotensin following its binding to the (pro)renin receptor. However, recent studies reported no evidence for prorenin expression in the brain, nor for its selective up-regulation in neurogenic hypertension, and the (pro)renin receptor rather displays RAS-unrelated functions. Finally, although angiotensinogen mRNA is detectable in the brain, brain angiotensinogen protein levels are low, and even these low levels might be an overestimation due to assay artefacts. Taken together, independent angiotensin generation in the brain is unlikely. Indeed, brain angiotensin levels are extremely low, with angiotensin (Ang) I levels corresponding to the small amounts of Ang I in trapped blood plasma, and Ang II levels at most representing Ang II bound to (vascular) brain Ang II type 1 receptors. This review concludes with a unifying concept proposing the blood origin of angiotensin in the brain, possibly resulting in increased levels following blood-brain barrier disruption (e.g. due to hypertension), and suggesting that interfering with either intracellular renin or the (pro)renin receptor has consequences in an RAS-independent manner.
Collapse
|
46
|
Parrish DC, Francis Stuart SD, Olivas A, Wang L, Nykjaer A, Ripplinger CM, Habecker BA. Transient denervation of viable myocardium after myocardial infarction does not alter arrhythmia susceptibility. Am J Physiol Heart Circ Physiol 2018; 314:H415-H423. [PMID: 29101167 PMCID: PMC5899257 DOI: 10.1152/ajpheart.00300.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 10/30/2017] [Accepted: 10/30/2017] [Indexed: 01/14/2023]
Abstract
Cardiac sympathetic nerves stimulate heart rate and force of contraction. Myocardial infarction (MI) leads to the loss of sympathetic nerves within the heart, and clinical studies have indicated that sympathetic denervation is a risk factor for arrhythmias and cardiac arrest. Two distinct types of denervation have been identified in the mouse heart after MI caused by ischemia-reperfusion: transient denervation of peri-infarct myocardium and sustained denervation of the infarct. Sustained denervation is linked to increased arrhythmia risk, but it is not known whether acute nerve loss in peri-infarct myocardium also contributes to arrhythmia risk. Peri-infarct sympathetic denervation requires the p75 neurotrophin receptor (p75NTR), but removal of p75NTR alters the pattern of sympathetic innervation in the heart and increases spontaneous arrhythmias. Therefore, we targeted the p75NTR coreceptor sortilin and the p75NTR-induced protease tumor necrosis factor-α-converting enzyme/A disintegrin and metalloproteinase domain 17 (TACE/ADAM17) to selectively block peri-infarct denervation. Sympathetic nerve density was quantified using immunohistochemistry for tyrosine hydroxylase. Genetic deletion of sortilin had no effect on the timing or extent of axon degeneration, but inhibition of TACE/ADAM17 with the protease inhibitor marimastat prevented the loss of axons from viable myocardium. We then asked whether retention of nerves in peri-infarct myocardium had an impact on cardiac electrophysiology 3 days after MI using ex vivo optical mapping of transmembrane potential and intracellular Ca2+. Preventing acute denervation of viable myocardium after MI did not significantly alter cardiac electrophysiology or Ca2+ handling, suggesting that transient denervation at this early time point has minimal impact on arrhythmia risk. NEW & NOTEWORTHY Sympathetic denervation after myocardial infarction is a risk factor for arrhythmias. We asked whether transient loss of nerves in viable myocardium contributed to arrhythmia risk. We found that targeting protease activity could prevent acute peri-infarct denervation but that it did not significantly alter cardiac electrophysiology or Ca2+ handling 3 days after myocardial infarction.
Collapse
MESH Headings
- ADAM17 Protein/metabolism
- Action Potentials
- Adaptor Proteins, Vesicular Transport/metabolism
- Animals
- Arrhythmias, Cardiac/etiology
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/pathology
- Arrhythmias, Cardiac/physiopathology
- Calcium Signaling
- Disease Models, Animal
- Heart/innervation
- Heart Rate
- Isolated Heart Preparation
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardial Infarction/complications
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardial Infarction/physiopathology
- Myocardium/metabolism
- Myocardium/pathology
- Receptors, Nerve Growth Factor/deficiency
- Receptors, Nerve Growth Factor/genetics
- Sympathetic Nervous System/metabolism
- Sympathetic Nervous System/physiopathology
- Time Factors
- Tissue Survival
Collapse
Affiliation(s)
- Diana C Parrish
- Department of Physiology and Pharmacology, Oregon Health and Science University , Portland, Oregon
| | | | - Antoinette Olivas
- Department of Physiology and Pharmacology, Oregon Health and Science University , Portland, Oregon
| | - Lianguo Wang
- Department of Pharmacology, University of California , Davis, California
| | - Anders Nykjaer
- Department of Biomedicine-Medical Biochemistry, Aarhus University , Aarhus , Denmark
| | | | - Beth A Habecker
- Department of Physiology and Pharmacology, Oregon Health and Science University , Portland, Oregon
- Department of Medicine and Knight Cardiovascular Institute, Oregon Health and Science University , Portland, Oregon
| |
Collapse
|
47
|
Boggild S, Molgaard S, Glerup S, Nyengaard JR. Highly segregated localization of the functionally related vps10p receptors sortilin and SorCS2 during neurodevelopment. J Comp Neurol 2018; 526:1267-1286. [PMID: 29405286 DOI: 10.1002/cne.24403] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 01/20/2018] [Accepted: 01/22/2018] [Indexed: 12/11/2022]
Abstract
Nervous system development is a precisely orchestrated series of events requiring a multitude of intrinsic and extrinsic cues. Sortilin and SorCS2 are members of the Vps10p receptor family with complementary influence on some of these cues including the neurotrophins (NTs). However, the developmental time points where sortilin and SorCS2 exert their activities in conjunction or independently still remain unclear. In this study we present the characterization of the spatiotemporal expression pattern of sortilin and SorCS2 in the developing murine nervous system. Sortilin is highly expressed in the fetal nervous system with expression localized to distinct cell populations. Expression was high in neurons of the cortical plate and developing allocortex, as well as subpallial structures. Furthermore, the neuroepithelium lining the ventricles and the choroid plexus showed high expression of sortilin, together with the developing retina, spinal ganglia, and sympathetic ganglia. In contrast, SorCS2 was confined in a marked degree to the thalamus and, at E13.5, the floor plate from midbrain rostrally to spinal cord caudally. SorCS2 was also found in the ventricular zones of the ventral hippocampus and nucleus accumbens areas, in the meninges and in Schwann cells. Hence, sortilin and SorCS2 are extensively present in several distinct anatomical areas in the developing nervous system and are rarely co-expressed. Possible functions of sortilin and SorCS2 pertain to NT signaling, axon guidance and beyond. The present data will form the basis for hypotheses and study designs for unravelling the functions of sortilin and SorCS2 during the establishment of neuronal structures and connections.
Collapse
Affiliation(s)
- Simon Boggild
- Department of Clinical Medicine, Aarhus University, MIND Centre, Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus C, 8000, Denmark.,MIND Centre, Department of Biomedicine, Aarhus University, Ole Worms Allé 3, Aarhus C, 8000, Denmark
| | - Simon Molgaard
- Department of Clinical Medicine, Aarhus University, MIND Centre, Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus C, 8000, Denmark.,MIND Centre, Department of Biomedicine, Aarhus University, Ole Worms Allé 3, Aarhus C, 8000, Denmark
| | - Simon Glerup
- MIND Centre, Department of Biomedicine, Aarhus University, Ole Worms Allé 3, Aarhus C, 8000, Denmark
| | - Jens Randel Nyengaard
- Department of Clinical Medicine, Aarhus University, MIND Centre, Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus C, 8000, Denmark.,Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus C, 8000, Denmark
| |
Collapse
|
48
|
Johnson JJ, Loeffert AC, Stokes J, Olympia RP, Bramley H, Hicks SD. Association of Salivary MicroRNA Changes With Prolonged Concussion Symptoms. JAMA Pediatr 2018; 172:65-73. [PMID: 29159407 PMCID: PMC5833519 DOI: 10.1001/jamapediatrics.2017.3884] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
IMPORTANCE Approximately one-third of children who experience a concussion develop prolonged concussion symptoms. To our knowledge, there are currently no objective or easily administered tests for predicting prolonged concussion symptoms. Several studies have identified alterations in epigenetic molecules known as microRNAs (miRNAs) following traumatic brain injury. No studies have examined whether miRNA expression can detect prolonged concussion symptoms. OBJECTIVE To evaluate the efficacy of salivary miRNAs for identifying children with concussion who are at risk for prolonged symptoms. DESIGN, SETTING, AND PARTICIPANTS This prospective cohort study at the Penn State Medical Center observed 52 patients aged 7 to 21 years presenting for evaluation of concussion within 14 days of initial head injury, with follow-up at 4 and 8 weeks. EXPOSURES All patients had a clinical diagnosis of concussion. MAIN OUTCOMES AND MEASURES Salivary miRNA expression was measured at the time of initial clinical presentation in all patients. Patients with a Sport Concussion Assessment Tool (SCAT3) symptom score of 5 or greater on self-report or parent report 4 weeks after injury were designated as having prolonged symptoms. RESULTS Of the 52 included participants, 22 (42%) were female, and the mean (SD) age was 14 (3) years. Participants were split into the prolonged symptom group (n = 30) and acute symptom group (n = 22). Concentrations of 15 salivary miRNAs spatially differentiated prolonged and acute symptom groups on partial least squares discriminant analysis and demonstrated functional relationships with neuronal regulatory pathways. Levels of 5 miRNAs (miR-320c-1, miR-133a-5p, miR-769-5p, let-7a-3p, and miR-1307-3p) accurately identified patients with prolonged symptoms on logistic regression (area under the curve, 0.856; 95% CI, 0.822-0.890). This accuracy exceeded accuracy of symptom burden on child (area under the curve, 0.649; 95% CI, 0.388-0.887) or parent (area under the curve, 0.562; 95% CI, 0.219-0.734) SCAT3 score. Levels of 3 miRNAs were associated with specific symptoms 4 weeks after injury; miR-320c-1 was associated with memory difficulty (R, 0.55; false detection rate, 0.02), miR-629 was associated with headaches (R, 0.47; false detection rate, 0.04), and let-7b-5p was associated with fatigue (R, 0.45; false detection rate, 0.04). CONCLUSIONS AND RELEVANCE Salivary miRNA levels may identify the duration and character of concussion symptoms. This could reduce parental anxiety and improve care by providing a tool for concussion management. Further validation of this approach is needed.
Collapse
Affiliation(s)
- Jeremiah J. Johnson
- Department of Pediatrics, Penn State College of Medicine, Pennsylvania State University, Hershey
| | - Andrea C. Loeffert
- Department of Pediatrics, Penn State College of Medicine, Pennsylvania State University, Hershey
| | - Jennifer Stokes
- Department of Pediatrics, Penn State College of Medicine, Pennsylvania State University, Hershey
| | - Robert P. Olympia
- Department of Pediatrics, Penn State College of Medicine, Pennsylvania State University, Hershey,Department of Emergency Medicine, Penn State College of Medicine, Pennsylvania State University, Hershey
| | - Harry Bramley
- Department of Pediatrics, Penn State College of Medicine, Pennsylvania State University, Hershey
| | - Steven D. Hicks
- Department of Pediatrics, Penn State College of Medicine, Pennsylvania State University, Hershey
| |
Collapse
|
49
|
Schwarz E. Cystine knot growth factors and their functionally versatile proregions. Biol Chem 2017; 398:1295-1308. [PMID: 28771427 DOI: 10.1515/hsz-2017-0163] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/16/2017] [Indexed: 12/23/2022]
Abstract
The cystine knot disulfide pattern has been found to be widespread in nature, since it has been detected in proteins from plants, marine snails, spiders and mammals. Cystine knot proteins are secreted proteins. Their functions range from defense mechanisms as toxins, e.g. ion channel or enzyme inhibitors, to hormones, blood factors and growth factors. Cystine knot proteins can be divided into two superordinate groups. (i) The cystine knot peptides, also referred to - with other non-cystine knot proteins - as knottins, with linear and cyclic polypeptide chains. (ii) The cystine knot growth factor family, which is in the focus of this article. The disulfide ring structure of the cystine knot peptides is made up by the half-cystines 1-4 and 2-5, and the threading disulfide bond is formed by the half-cystines, 3-6. In the growth factor group, the disulfides of half-cystines 1 and 4 pass the ring structure formed by the half-cystines 2-5 and 3-6. In this review, special emphasis will be devoted to the growth factor cystine knot proteins and their proregions. The latter have shifted into the focus of scientific interest as their important biological roles are just to be unravelled.
Collapse
|
50
|
Trabjerg E, Kartberg F, Christensen S, Rand KD. Conformational characterization of nerve growth factor-β reveals that its regulatory pro-part domain stabilizes three loop regions in its mature part. J Biol Chem 2017; 292:16665-16676. [PMID: 28798232 DOI: 10.1074/jbc.m117.803320] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/07/2017] [Indexed: 11/06/2022] Open
Abstract
Nerve growth factor-β (NGF) is essential for the correct development of the nervous system. NGF exists in both a mature form and a pro-form (proNGF). The two forms have opposing effects on neurons: NGF induces proliferation, whereas proNGF induces apoptosis via binding to a receptor complex of the common neurotrophin receptor (p75NTR) and sortilin. The overexpression of both proNGF and sortilin has been associated with several neurodegenerative diseases. Insights into the conformational differences between proNGF and NGF are central to a better understanding of the opposing mechanisms of action of NGF and proNGF on neurons. However, whereas the structure of NGF has been determined by X-ray crystallography, the structural details for proNGF remain elusive. Here, using a sensitive MS-based analytical method to measure the hydrogen/deuterium exchange of proteins in solution, we analyzed the conformational properties of proNGF and NGF. We detected the presence of a localized higher-order structure motif in the pro-part of proNGF. Furthermore, by comparing the hydrogen/deuterium exchange in the mature part of NGF and proNGF, we found that the presence of the pro-part in proNGF causes a structural stabilization of three loop regions in the mature part, possibly through a direct molecular interaction. Moreover, using tandem MS analyses, we identified two N-linked and two O-linked glycosylations in the pro-part of proNGF. These results advance our knowledge of the conformational properties of proNGF and NGF and help provide a rationale for the diverse biological effects of NGF and proNGF at the molecular level.
Collapse
Affiliation(s)
- Esben Trabjerg
- From the Department of Pharmacy, University of Copenhagen, 2100 Copenhagen E, Denmark and.,the Department of Biologics, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Fredrik Kartberg
- the Department of Biologics, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Søren Christensen
- the Department of Biologics, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Kasper D Rand
- From the Department of Pharmacy, University of Copenhagen, 2100 Copenhagen E, Denmark and
| |
Collapse
|