1
|
Han X, Zhang M, Yan L, Fu Y, Kou H, Shang C, Wang J, Liu H, Jiang C, Wang J, Cheng T. Role of dendritic cells in spinal cord injury. CNS Neurosci Ther 2024; 30:e14593. [PMID: 38528832 PMCID: PMC10964036 DOI: 10.1111/cns.14593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/15/2023] [Accepted: 12/10/2023] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Inflammation can worsen spinal cord injury (SCI), with dendritic cells (DCs) playing a crucial role in the inflammatory response. They mediate T lymphocyte differentiation, activate microglia, and release cytokines like NT-3. Moreover, DCs can promote neural stem cell survival and guide them toward neuron differentiation, positively impacting SCI outcomes. OBJECTIVE This review aims to summarize the role of DCs in SCI-related inflammation and identify potential therapeutic targets for treating SCI. METHODS Literature in PubMed and Web of Science was reviewed using critical terms related to DCs and SCI. RESULTS The study indicates that DCs can activate microglia and astrocytes, promote T-cell differentiation, increase neurotrophin release at the injury site, and subsequently reduce secondary brain injury and enhance functional recovery in the spinal cord. CONCLUSIONS This review highlights the repair mechanisms of DCs and their potential therapeutic potential for SCI.
Collapse
Affiliation(s)
- Xiaonan Han
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Mingkang Zhang
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Liyan Yan
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yikun Fu
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hongwei Kou
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Chunfeng Shang
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Junmin Wang
- Department of Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Hongjian Liu
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Chao Jiang
- Department of NeurologyThe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jian Wang
- Department of Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Tian Cheng
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
2
|
Zhao L, Hou C, Yan N. Neuroinflammation in retinitis pigmentosa: Therapies targeting the innate immune system. Front Immunol 2022; 13:1059947. [PMID: 36389729 PMCID: PMC9647059 DOI: 10.3389/fimmu.2022.1059947] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
Retinitis pigmentosa (RP) is an important cause of irreversible blindness worldwide and lacks effective treatment strategies. Although mutations are the primary cause of RP, research over the past decades has shown that neuroinflammation is an important cause of RP progression. Due to the abnormal activation of immunity, continuous sterile inflammation results in neuron loss and structural destruction. Therapies targeting inflammation have shown their potential to attenuate photoreceptor degeneration in preclinical models. Regardless of variations in genetic background, inflammatory modulation is emerging as an important role in the treatment of RP. We summarize the evidence for the role of inflammation in RP and mention therapeutic strategies where available, focusing on the modulation of innate immune signals, including TNFα signaling, TLR signaling, NLRP3 inflammasome activation, chemokine signaling and JAK/STAT signaling. In addition, we describe epigenetic regulation, the gut microbiome and herbal agents as prospective treatment strategies for RP in recent advances.
Collapse
Affiliation(s)
- Ling Zhao
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Hou
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Naihong Yan
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Naihong Yan,
| |
Collapse
|
3
|
Human-Induced Pluripotent Stem Cell-Based Models for Studying Sex-Specific Differences in Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1387:57-88. [PMID: 34921676 DOI: 10.1007/5584_2021_683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The prevalence of neurodegenerative diseases is steadily increasing worldwide, and epidemiological studies strongly suggest that many of the diseases are sex-biased. It has long been suggested that biological sex differences are crucial for neurodegenerative diseases; however, how biological sex affects disease initiation, progression, and severity is not well-understood. Sex is a critical biological variable that should be taken into account in basic research, and this review aims to highlight the utility of human-induced pluripotent stem cells (iPSC)-derived models for studying sex-specific differences in neurodegenerative diseases, with advantages and limitations. In vitro systems utilizing species-specific, renewable, and physiologically relevant cell sources can provide powerful platforms for mechanistic studies, toxicity testings, and drug discovery. Matched healthy, patient-derived, and gene-corrected human iPSCs, from both sexes, can be utilized to generate neuronal and glial cell types affected by specific neurodegenerative diseases to study sex-specific differences in two-dimensional (2D) and three-dimensional (3D) human culture systems. Such relatively simple and well-controlled systems can significantly contribute to the elucidation of molecular mechanisms underlying sex-specific differences, which can yield effective, and potentially sex-based strategies, against neurodegenerative diseases.
Collapse
|
4
|
Brahimi F, Galan A, Siegel S, Szobota S, Sarunic MV, Foster AC, Saragovi HU. Therapeutic Neuroprotection by an Engineered Neurotrophin that Selectively Activates Tropomyosin Receptor Kinase (Trk) Family Neurotrophin Receptors but Not the p75 Neurotrophin Receptor. Mol Pharmacol 2021; 100:491-501. [PMID: 34470776 DOI: 10.1124/molpharm.121.000301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/12/2021] [Indexed: 11/22/2022] Open
Abstract
The neurotrophin growth factors bind and activate two types of cell surface receptors: the tropomyosin receptor kinase (Trk) family and p75. TrkA, TrkB, and TrkC are bound preferentially by nerve growth factor, brain-derived neurotrophic factor, and neurotrophin 3 (NT3), respectively, to activate neuroprotective signals. The p75 receptors are activated by all neurotrophins, and paradoxically in neurodegenerative disease p75 is upregulated and mediates neurotoxic signals. To test neuroprotection strategies, we engineered NT3 to broadly activate Trk receptors (mutant D) or to reduce p75 binding (mutant RK). We also combined these features in a molecule that activates TrkA, TrkB, and TrkC but has reduced p75 binding (mutant DRK). In neurodegenerative disease mouse models in vivo, the DRK protein is a superior therapeutic agent compared with mutant D, mutant RK, and wild-type neurotrophins and protects a broader range of stressed neurons. This work rationalizes a therapeutic strategy based on the biology of each type of receptor, avoiding activation of p75 toxicity while broadly activating neuroprotection in stressed neuronal populations expressing different Trk receptors. SIGNIFICANCE STATEMENT: The neurotrophins nerve growth factor, brain-derived neurotrophic factor, and neurotrophin 3 each can activate a tropomyosin receptor kinase (Trk) A, TrkB, or TrkC receptor, respectively, and all can activate a p75 receptor. Trks and p75 mediate opposite signals. We report the engineering of a protein that activates all Trks, combined with low p75 binding, as an effective therapeutic agent in vivo.
Collapse
Affiliation(s)
- Fouad Brahimi
- Lady Davis Institute-Jewish General Hospital (F.B., A.G., H.U.S.), Pharmacology and Therapeutics (H.U.S.), and Ophthalmology and Vision Science (H.U.S.), McGill University, Montreal, Quebec, Canada; Otonomy, Inc., San Diego, California (S.Si., S.Sz., A.C.F.); and School of Engineering Science, Simon Fraser University, British Columbia, Canada (M.V.S.)
| | - Alba Galan
- Lady Davis Institute-Jewish General Hospital (F.B., A.G., H.U.S.), Pharmacology and Therapeutics (H.U.S.), and Ophthalmology and Vision Science (H.U.S.), McGill University, Montreal, Quebec, Canada; Otonomy, Inc., San Diego, California (S.Si., S.Sz., A.C.F.); and School of Engineering Science, Simon Fraser University, British Columbia, Canada (M.V.S.)
| | - Sairey Siegel
- Lady Davis Institute-Jewish General Hospital (F.B., A.G., H.U.S.), Pharmacology and Therapeutics (H.U.S.), and Ophthalmology and Vision Science (H.U.S.), McGill University, Montreal, Quebec, Canada; Otonomy, Inc., San Diego, California (S.Si., S.Sz., A.C.F.); and School of Engineering Science, Simon Fraser University, British Columbia, Canada (M.V.S.)
| | - Stephanie Szobota
- Lady Davis Institute-Jewish General Hospital (F.B., A.G., H.U.S.), Pharmacology and Therapeutics (H.U.S.), and Ophthalmology and Vision Science (H.U.S.), McGill University, Montreal, Quebec, Canada; Otonomy, Inc., San Diego, California (S.Si., S.Sz., A.C.F.); and School of Engineering Science, Simon Fraser University, British Columbia, Canada (M.V.S.)
| | - Marinko V Sarunic
- Lady Davis Institute-Jewish General Hospital (F.B., A.G., H.U.S.), Pharmacology and Therapeutics (H.U.S.), and Ophthalmology and Vision Science (H.U.S.), McGill University, Montreal, Quebec, Canada; Otonomy, Inc., San Diego, California (S.Si., S.Sz., A.C.F.); and School of Engineering Science, Simon Fraser University, British Columbia, Canada (M.V.S.)
| | - Alan C Foster
- Lady Davis Institute-Jewish General Hospital (F.B., A.G., H.U.S.), Pharmacology and Therapeutics (H.U.S.), and Ophthalmology and Vision Science (H.U.S.), McGill University, Montreal, Quebec, Canada; Otonomy, Inc., San Diego, California (S.Si., S.Sz., A.C.F.); and School of Engineering Science, Simon Fraser University, British Columbia, Canada (M.V.S.)
| | - H Uri Saragovi
- Lady Davis Institute-Jewish General Hospital (F.B., A.G., H.U.S.), Pharmacology and Therapeutics (H.U.S.), and Ophthalmology and Vision Science (H.U.S.), McGill University, Montreal, Quebec, Canada; Otonomy, Inc., San Diego, California (S.Si., S.Sz., A.C.F.); and School of Engineering Science, Simon Fraser University, British Columbia, Canada (M.V.S.)
| |
Collapse
|
5
|
Dedoni S, Marras L, Olianas MC, Ingianni A, Onali P. The Neurotrophin Receptor TrkC as a Novel Molecular Target of the Antineuroblastoma Action of Valproic Acid. Int J Mol Sci 2021; 22:ijms22157790. [PMID: 34360553 PMCID: PMC8346142 DOI: 10.3390/ijms22157790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/14/2021] [Accepted: 07/19/2021] [Indexed: 11/16/2022] Open
Abstract
Neurotrophins and their receptors are relevant factors in controlling neuroblastoma growth and progression. The histone deacetylase (HDAC) inhibitor valproic acid (VPA) has been shown to downregulate TrkB and upregulate the p75NTR/sortilin receptor complex. In the present study, we investigated the VPA effect on the expression of the neurotrophin-3 (NT-3) receptor TrkC, a favorable prognostic marker of neuroblastoma. We found that VPA induced the expression of both full-length and truncated (TrkC-T1) isoforms of TrkC in human neuroblastoma cell lines without (SH-SY5Y) and with (Kelly, BE(2)-C and IMR 32) MYCN amplification. VPA enhanced cell surface expression of the receptor and increased Akt and ERK1/2 activation by NT-3. The HDAC inhibitors entinostat, romidepsin and vorinostat also increased TrkC in SH-SY5Y, Kelly and BE(2)-C but not IMR 32 cells. TrkC upregulation by VPA involved induction of RUNX3, stimulation of ERK1/2 and JNK, and ERK1/2-mediated Egr1 expression. In SH-SY5Y cell monolayers and spheroids the exposure to NT-3 enhanced the apoptotic cascade triggered by VPA. Gene silencing of both TrkC-T1 and p75NTR prevented the NT-3 proapoptotic effect. Moreover, NT-3 enhanced p75NTR/TrkC-T1 co-immunoprecipitation. The results indicate that VPA upregulates TrkC by activating epigenetic mechanisms and signaling pathways, and sensitizes neuroblastoma cells to NT-3-induced apoptosis.
Collapse
Affiliation(s)
- Simona Dedoni
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, University of Cagliari, 09042 Monserrato, Italy; (S.D.); (M.C.O.)
| | - Luisa Marras
- Section of Microbiology, Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (L.M.); (A.I.)
| | - Maria C. Olianas
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, University of Cagliari, 09042 Monserrato, Italy; (S.D.); (M.C.O.)
| | - Angela Ingianni
- Section of Microbiology, Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (L.M.); (A.I.)
| | - Pierluigi Onali
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, University of Cagliari, 09042 Monserrato, Italy; (S.D.); (M.C.O.)
- Correspondence:
| |
Collapse
|
6
|
Li G, Zhang B, Sun JH, Shi LY, Huang MY, Huang LJ, Lin ZJ, Lin QY, Lai BQ, Ma YH, Jiang B, Ding Y, Zhang HB, Li MX, Zhu P, Wang YQ, Zeng X, Zeng YS. An NT-3-releasing bioscaffold supports the formation of TrkC-modified neural stem cell-derived neural network tissue with efficacy in repairing spinal cord injury. Bioact Mater 2021; 6:3766-3781. [PMID: 33898877 PMCID: PMC8044869 DOI: 10.1016/j.bioactmat.2021.03.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/03/2021] [Accepted: 03/18/2021] [Indexed: 01/01/2023] Open
Abstract
The mechanism underlying neurogenesis during embryonic spinal cord development involves a specific ligand/receptor interaction, which may be help guide neuroengineering to boost stem cell-based neural regeneration for the structural and functional repair of spinal cord injury. Herein, we hypothesized that supplying spinal cord defects with an exogenous neural network in the NT-3/fibroin-coated gelatin sponge (NF-GS) scaffold might improve tissue repair efficacy. To test this, we engineered tropomyosin receptor kinase C (TrkC)-modified neural stem cell (NSC)-derived neural network tissue with robust viability within an NF-GS scaffold. When NSCs were genetically modified to overexpress TrkC, the NT-3 receptor, a functional neuronal population dominated the neural network tissue. The pro-regenerative niche allowed the long-term survival and phenotypic maintenance of the donor neural network tissue for up to 8 weeks in the injured spinal cord. Additionally, host nerve fibers regenerated into the graft, making synaptic connections with the donor neurons. Accordingly, motor function recovery was significantly improved in rats with spinal cord injury (SCI) that received TrkC-modified NSC-derived neural network tissue transplantation. Together, the results suggested that transplantation of the neural network tissue formed in the 3D bioactive scaffold may represent a valuable approach to study and develop therapies for SCI. A NT-3 sustained-release scaffold confers a microenvironment partially simulating the developmental spinal cord. The NT-3 microenvironment boosts neuronal differentiation of TrkC-modified NSCs by interactions between ligand and receptor. TrkC-NSCs is self-organized into a neural network tissue with typical neural excitability in 3D bioactive scaffold in vitro. The grafted neural network tissue can survive and maintain neural property, and improve motor function of paralyzed rats.
Collapse
Affiliation(s)
- Ge Li
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China
| | - Bao Zhang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jia-Hui Sun
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Li-Yang Shi
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, 410082, China
| | - Meng-Yao Huang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Li-Jun Huang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zi-Jing Lin
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qiong-Yu Lin
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China
| | - Bi-Qin Lai
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yuan-Huan Ma
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China
| | - Bin Jiang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ying Ding
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China
| | - Hong-Bo Zhang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Miao-Xin Li
- Laboratory of Precision Medical Genomics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China
| | - Ya-Qiong Wang
- Department of Electron Microscope, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yuan-Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
7
|
Brahimi F, Galan A, Jmaeff S, Barcelona PF, De Jay N, Dejgaard K, Young JC, Kleinman CL, Thomas DY, Saragovi HU. Alternative Splicing of a Receptor Intracellular Domain Yields Different Ectodomain Conformations, Enabling Isoform-Selective Functional Ligands. iScience 2020; 23:101447. [PMID: 32829283 PMCID: PMC7452315 DOI: 10.1016/j.isci.2020.101447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/13/2020] [Accepted: 08/06/2020] [Indexed: 01/04/2023] Open
Abstract
Events at a receptor ectodomain affect the intracellular domain conformation, activating signal transduction (out-to-in conformational effects). We investigated the reverse direction (in-to-out) where the intracellular domain may impact on ectodomain conformation. The primary sequences of naturally occurring TrkC receptor isoforms (TrkC-FL and TrkC.T1) only differ at the intracellular domain. However, owing to their differential association with Protein Disulfide Isomerase the isoforms have different disulfide bonding and conformations at the ectodomain. Conformations were exploited to develop artificial ligands, mAbs, and small molecules, with isoform-specific binding and biased activation. Consistent, the physiological ligands NT-3 and PTP-sigma bind both isoforms, but NT-3 activates all signaling pathways, whereas PTP-sigma activates biased signals. Our data support an "in-to-out" model controlling receptor ectodomain conformation, a strategy that enables heterogeneity in receptors, ligands, and bioactivity. These concepts may be extended to the many wild-type or oncogenic receptors with known isoforms.
Collapse
Affiliation(s)
- Fouad Brahimi
- Lady Davis Institute-Jewish General Hospital, McGill University, 3755 Côte St. Catherine, E-535, Montreal, QC H3T 1E2, Canada
| | - Alba Galan
- Lady Davis Institute-Jewish General Hospital, McGill University, 3755 Côte St. Catherine, E-535, Montreal, QC H3T 1E2, Canada
| | - Sean Jmaeff
- Lady Davis Institute-Jewish General Hospital, McGill University, 3755 Côte St. Catherine, E-535, Montreal, QC H3T 1E2, Canada
- Department of Pharmacology, McGill University, Montreal, QC, Canada
| | - Pablo F. Barcelona
- Lady Davis Institute-Jewish General Hospital, McGill University, 3755 Côte St. Catherine, E-535, Montreal, QC H3T 1E2, Canada
| | - Nicolas De Jay
- Lady Davis Institute-Jewish General Hospital, McGill University, 3755 Côte St. Catherine, E-535, Montreal, QC H3T 1E2, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Kurt Dejgaard
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Jason C. Young
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Claudia L. Kleinman
- Lady Davis Institute-Jewish General Hospital, McGill University, 3755 Côte St. Catherine, E-535, Montreal, QC H3T 1E2, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - David Y. Thomas
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - H. Uri Saragovi
- Lady Davis Institute-Jewish General Hospital, McGill University, 3755 Côte St. Catherine, E-535, Montreal, QC H3T 1E2, Canada
- Department of Pharmacology, McGill University, Montreal, QC, Canada
- Department of Ophthalmology and Visual Science, McGill University, Montreal, QC, Canada
| |
Collapse
|
8
|
Jmaeff S, Sidorova Y, Nedev H, Saarma M, Saragovi HU. Small-molecule agonists of the RET receptor tyrosine kinase activate biased trophic signals that are influenced by the presence of GFRa1 co-receptors. J Biol Chem 2020; 295:6532-6542. [PMID: 32245892 DOI: 10.1074/jbc.ra119.011802] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a growth factor that regulates the health and function of neurons and other cells. GDNF binds to GDNF family receptor α1 (GFRa1), and the resulting complex activates the RET receptor tyrosine kinase and subsequent downstream signals. This feature restricts GDNF activity to systems in which GFRa1 and RET are both present, a scenario that may constrain GDNF breadth of action. Furthermore, this co-dependence precludes the use of GDNF as a tool to study a putative functional cross-talk between GFRa1 and RET. Here, using biochemical techniques, terminal deoxynucleotidyl transferase dUTP nick end labeling staining, and immunohistochemistry in murine cells, tissues, or retinal organotypic cultures, we report that a naphthoquinone/quinolinedione family of small molecules (Q compounds) acts as RET agonists. We found that, like GDNF, signaling through the parental compound Q121 is GFRa1-dependent. Structural modifications of Q121 generated analogs that activated RET irrespective of GFRa1 expression. We used these analogs to examine RET-GFRa1 interactions and show that GFRa1 can influence RET-mediated signaling and enhance or diminish AKT Ser/Thr kinase or extracellular signal-regulated kinase signaling in a biased manner. In a genetic mutant model of retinitis pigmentosa, a lead compound, Q525, afforded sustained RET activation and prevented photoreceptor neuron loss in the retina. This work uncovers key components of the dynamic relationships between RET and its GFRa co-receptor and provides RET agonist scaffolds for drug development.
Collapse
Affiliation(s)
- Sean Jmaeff
- Lady Davis Institute-Jewish General Hospital, McGill University, Montreal, Quebec H3T 1E2, Canada.,Department of Pharmacology, McGill University, Montreal, Quebec H3T 1E2, Canada
| | - Yulia Sidorova
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| | - Hinyu Nedev
- Lady Davis Institute-Jewish General Hospital, McGill University, Montreal, Quebec H3T 1E2, Canada
| | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| | - H Uri Saragovi
- Lady Davis Institute-Jewish General Hospital, McGill University, Montreal, Quebec H3T 1E2, Canada .,Department of Pharmacology, McGill University, Montreal, Quebec H3T 1E2, Canada.,Department of Ophthalmology and Visual Science, McGill University, Montreal, Quebec H3T 1E2, Canada
| |
Collapse
|
9
|
Zhang X, Zhang R, Chen J, Wu J. Neuroprotective effects of DAAO are mediated via the ERK1/2 signaling pathway in a glaucomatous animal model. Exp Eye Res 2020; 190:107892. [DOI: 10.1016/j.exer.2019.107892] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/31/2019] [Accepted: 12/03/2019] [Indexed: 01/06/2023]
|
10
|
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: 3.2] [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
|