1
|
Pewklang T, Thompson T, Sefiani A, Geoffroy CG, Kamkaew A, Burgess K. Selective, Intrinsically Fluorescent Trk Modulating Probes. ACS Chem Neurosci 2024; 15. [PMID: 39356215 PMCID: PMC11487604 DOI: 10.1021/acschemneuro.4c00290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 10/03/2024] Open
Abstract
Neurotrophins (NTs) elicit the growth, survival, and differentiation of neurons and other neuroectoderm tissues via activation of Trk receptors. Hot spots for NT·Trk interactions involve three neurotrophin loops. Mimicry of these using "cyclo-organopeptides" comprising loop sequences cyclized onto endocyclic organic fragments accounts for a few of the low molecular mass Trk agonists or modulators reported so far; the majority are nonpeptidic small molecules accessed without molecular design and identified in random screens. It has proven difficult to verify activities induced by low molecular mass substances are due to Trk activation (rather than via other receptors), enhanced Trk expression, enhanced NT expression, or other pathways. Consequently, identification of selective probes for the various Trk receptors (e.g., A, B, and C) has been very challenging. Further, a key feature of probes for early stage assays is that they should be easily detectable, and none of the compounds reported to date are. In this work, we designed novel cyclo-organopeptide derivatives where the organic fragment is a BODIPY fluor and found ones that selectively, though not specifically, activate TrkA, B, or C. One of the assays used to reach this conclusion (binding to live Trk-expressing cells) relied on intrinsic fluorescence in the tested materials. Consequently, this work established low molecular mass Trk-selective probes exhibiting neuroprotective effects.
Collapse
Affiliation(s)
- Thitima Pewklang
- Department
of Chemistry, Texas A & M University, Box 30012, College Station, Texas 77842-3012, United States
- School
of Chemistry, Institute of Science, Suranaree
University of Technology, Nakhon
Ratchasima 30000, Thailand
| | - Tye Thompson
- Department
of Chemistry, Texas A & M University, Box 30012, College Station, Texas 77842-3012, United States
| | - Arthur Sefiani
- Department
of Neuroscience and Experimental Therapeutics, Texas A & M University Health Science Center, Bryan, Texas 77807, United States
- NeuroCreis,
Inc., College Station, Texas 77840, United States
| | - Cédric G. Geoffroy
- Department
of Neuroscience and Experimental Therapeutics, Texas A & M University Health Science Center, Bryan, Texas 77807, United States
- NeuroCreis,
Inc., College Station, Texas 77840, United States
| | - Anyanee Kamkaew
- School
of Chemistry, Institute of Science, Suranaree
University of Technology, Nakhon
Ratchasima 30000, Thailand
| | - Kevin Burgess
- Department
of Chemistry, Texas A & M University, Box 30012, College Station, Texas 77842-3012, United States
| |
Collapse
|
2
|
Zhao S, Mo G, Wang Q, Xu J, Yu S, Huang Z, Liu W, Zhang W. Role of RB1 in neurodegenerative diseases: inhibition of post-mitotic neuronal apoptosis via Kmt5b. Cell Death Discov 2024; 10:182. [PMID: 38637503 PMCID: PMC11026443 DOI: 10.1038/s41420-024-01955-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/04/2023] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
During the development of the vertebrate nervous system, 50% of the nerve cells undergo apoptosis shortly after formation. This process is important for sculpting tissue during morphogenesis and removing transiently functional cells that are no longer needed, ensuring the appropriate number of neurons in each region. Dysregulation of neuronal apoptosis can lead to neurodegenerative diseases. However, the molecular events involved in activating and regulating the neuronal apoptosis program are not fully understood. In this study, we identified several RB1 mutations in patients with neurodegenerative diseases. Then, we used a zebrafish model to investigate the role of Rb1 in neuronal apoptosis. We showed that Rb1-deficient mutants exhibit a significant hindbrain neuronal apoptosis, resulting in increased microglia infiltration. We further revealed that the apoptotic neurons in Rb1-deficient zebrafish were post-mitotic neurons, and Rb1 inhibits the apoptosis of these neurons by regulating bcl2/caspase through binding to Kmt5b. Moreover, using this zebrafish mutant, we verified the pathogenicity of the R621S and L819V mutations of human RB1 in neuronal apoptosis. Collectively, our data indicate that the Rb1-Kmt5b-caspase/bcl2 axis is crucial for protecting post-mitotic neurons from apoptosis and provides an explanation for the pathogenesis of clinically relevant mutations.
Collapse
Affiliation(s)
- Shuang Zhao
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Guiling Mo
- Guangzhou KingMed Diagnostics Group Co., Ltd., International Biotech Island, Guangzhou, 510005, China
| | - Qiang Wang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Jin Xu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Shihui Yu
- Guangzhou KingMed Diagnostics Group Co., Ltd., International Biotech Island, Guangzhou, 510005, China
| | - Zhibin Huang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Wei Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| | - Wenqing Zhang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
3
|
Paşcalău R, Badea TC. Signaling - transcription interactions in mouse retinal ganglion cells early axon pathfinding -a literature review. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1180142. [PMID: 38983012 PMCID: PMC11182120 DOI: 10.3389/fopht.2023.1180142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 04/21/2023] [Indexed: 07/11/2024]
Abstract
Sending an axon out of the eye and into the target brain nuclei is the defining feature of retinal ganglion cells (RGCs). The literature on RGC axon pathfinding is vast, but it focuses mostly on decision making events such as midline crossing at the optic chiasm or retinotopic mapping at the target nuclei. In comparison, the exit of RGC axons out of the eye is much less explored. The first checkpoint on the RGC axons' path is the optic cup - optic stalk junction (OC-OS). OC-OS development and the exit of the RGC pioneer axons out of the eye are coordinated spatially and temporally. By the time the optic nerve head domain is specified, the optic fissure margins are in contact and the fusion process is ongoing, the first RGCs are born in its proximity and send pioneer axons in the optic stalk. RGC differentiation continues in centrifugal waves. Later born RGC axons fasciculate with the more mature axons. Growth cones at the end of the axons respond to guidance cues to adopt a centripetal direction, maintain nerve fiber layer restriction and to leave the optic cup. Although there is extensive information on OC-OS development, we still have important unanswered questions regarding its contribution to the exit of the RGC axons out of the eye. We are still to distinguish the morphogens of the OC-OS from the axon guidance molecules which are expressed in the same place at the same time. The early RGC transcription programs responsible for axon emergence and pathfinding are also unknown. This review summarizes the molecular mechanisms for early RGC axon guidance by contextualizing mouse knock-out studies on OC-OS development with the recent transcriptomic studies on developing RGCs in an attempt to contribute to the understanding of human optic nerve developmental anomalies. The published data summarized here suggests that the developing optic nerve head provides a physical channel (the closing optic fissure) as well as molecular guidance cues for the pioneer RGC axons to exit the eye.
Collapse
Affiliation(s)
- Raluca Paşcalău
- Research and Development Institute, Transilvania University of Braşov, Braşov, Romania
- Ophthalmology Clinic, Cluj County Emergency Hospital, Cluj-Napoca, Romania
| | - Tudor Constantin Badea
- Research and Development Institute, Transilvania University of Braşov, Braşov, Romania
- National Center for Brain Research, Institutul de Cercetări pentru Inteligență Artificială, Romanian Academy, Bucharest, Romania
| |
Collapse
|
4
|
Murenu E, Gerhardt MJ, Biel M, Michalakis S. More than meets the eye: The role of microglia in healthy and diseased retina. Front Immunol 2022; 13:1006897. [PMID: 36524119 PMCID: PMC9745050 DOI: 10.3389/fimmu.2022.1006897] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/11/2022] [Indexed: 11/30/2022] Open
Abstract
Microglia are the main resident immune cells of the nervous system and as such they are involved in multiple roles ranging from tissue homeostasis to response to insults and circuit refinement. While most knowledge about microglia comes from brain studies, some mechanisms have been confirmed for microglia cells in the retina, the light-sensing compartment of the eye responsible for initial processing of visual information. However, several key pieces of this puzzle are still unaccounted for, as the characterization of retinal microglia has long been hindered by the reduced population size within the retina as well as the previous lack of technologies enabling single-cell analyses. Accumulating evidence indicates that the same cell type may harbor a high degree of transcriptional, morphological and functional differences depending on its location within the central nervous system. Thus, studying the roles and signatures adopted specifically by microglia in the retina has become increasingly important. Here, we review the current understanding of retinal microglia cells in physiology and in disease, with particular emphasis on newly discovered mechanisms and future research directions.
Collapse
Affiliation(s)
- Elisa Murenu
- Department of Ophthalmology, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany,*Correspondence: Elisa Murenu, ; ; Stylianos Michalakis,
| | | | - Martin Biel
- Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stylianos Michalakis
- Department of Ophthalmology, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany,*Correspondence: Elisa Murenu, ; ; Stylianos Michalakis,
| |
Collapse
|
5
|
Fitzpatrick MJ, Kerschensteiner D. Homeostatic plasticity in the retina. Prog Retin Eye Res 2022; 94:101131. [PMID: 36244950 DOI: 10.1016/j.preteyeres.2022.101131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 02/07/2023]
Abstract
Vision begins in the retina, whose intricate neural circuits extract salient features of the environment from the light entering our eyes. Neurodegenerative diseases of the retina (e.g., inherited retinal degenerations, age-related macular degeneration, and glaucoma) impair vision and cause blindness in a growing number of people worldwide. Increasing evidence indicates that homeostatic plasticity (i.e., the drive of a neural system to stabilize its function) can, in principle, preserve retinal function in the face of major perturbations, including neurodegeneration. Here, we review the circumstances and events that trigger homeostatic plasticity in the retina during development, sensory experience, and disease. We discuss the diverse mechanisms that cooperate to compensate and the set points and outcomes that homeostatic retinal plasticity stabilizes. Finally, we summarize the opportunities and challenges for unlocking the therapeutic potential of homeostatic plasticity. Homeostatic plasticity is fundamental to understanding retinal development and function and could be an important tool in the fight to preserve and restore vision.
Collapse
|
6
|
Thiry L, Clément JP, Haag R, Kennedy TE, Stifani S. Optimization of Long-Term Human iPSC-Derived Spinal Motor Neuron Culture Using a Dendritic Polyglycerol Amine-Based Substrate. ASN Neuro 2022; 14:17590914211073381. [PMID: 35023784 PMCID: PMC8784909 DOI: 10.1177/17590914211073381] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/14/2021] [Accepted: 12/22/2021] [Indexed: 11/30/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) derived from healthy and diseased individuals can give rise to many cell types, facilitating the study of mechanisms of development, human disease modeling, and early drug target validation. In this context, experimental model systems based on hiPSC-derived motor neurons (MNs) have been used to study MN diseases such as spinal muscular atrophy and amyotrophic lateral sclerosis. Modeling MN disease using hiPSC-based approaches requires culture conditions that can recapitulate in a dish the events underlying differentiation, maturation, aging, and death of MNs. Current hiPSC-derived MN-based applications are often hampered by limitations in our ability to monitor MN morphology, survival, and other functional properties over a prolonged timeframe, underscoring the need for improved long-term culture conditions. Here we describe a cytocompatible dendritic polyglycerol amine (dPGA) substrate-based method for prolonged culture of hiPSC-derived MNs. We provide evidence that MNs cultured on dPGA-coated dishes are more amenable to long-term study of cell viability, molecular identity, and spontaneous network electrophysiological activity. The present study has the potential to improve hiPSC-based studies of human MN biology and disease.We describe the use of a new coating substrate providing improved conditions for long-term cultures of human iPSC-derived motor neurons, thus allowing evaluation of cell viability, molecular identity, spontaneous network electrophysiological activity, and single-cell RNA sequencing of mature motor neurons.
Collapse
Affiliation(s)
- Louise Thiry
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada
| | - Jean-Pierre Clément
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Timothy E Kennedy
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada
| | - Stefano Stifani
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada
| |
Collapse
|
7
|
Deshwal A, Phan P, Datta J, Kannan R, Thallapuranam SK. A Meta-Analysis of the Protein Components in Rattlesnake Venom. Toxins (Basel) 2021; 13:toxins13060372. [PMID: 34071038 DOI: 10.3390/toxins13060372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 11/16/2022] Open
Abstract
The specificity and potency of venom components give them a unique advantage in developing various pharmaceutical drugs. Though venom is a cocktail of proteins, rarely are the synergy and association between various venom components studied. Understanding the relationship between various components of venom is critical in medical research. Using meta-analysis, we observed underlying patterns and associations in the appearance of the toxin families. For Crotalus, Dis has the most associations with the following toxins: PDE; BPP; CRL; CRiSP; LAAO; SVMP P-I and LAAO; SVMP P-III and LAAO. In Sistrurus venom, CTL and NGF have the most associations. These associations can predict the presence of proteins in novel venom and understand synergies between venom components for enhanced bioactivity. Using this approach, the need to revisit the classification of proteins as major components or minor components is highlighted. The revised classification of venom components is based on ubiquity, bioactivity, the number of associations, and synergies. The revised classification can be expected to trigger increased research on venom components, such as NGF, which have high biomedical significance. Using hierarchical clustering, we observed that the genera's venom compositions were similar, based on functional characteristics rather than phylogenetic relationships.
Collapse
Affiliation(s)
- Anant Deshwal
- Division of Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Phuc Phan
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA
| | - Jyotishka Datta
- Department of Statistics, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Ragupathy Kannan
- Department of Biology, University of Arkansas-Fort Smith, Fort Smith, AR 72913, USA
| | | |
Collapse
|
8
|
Thiry L, Hamel R, Pluchino S, Durcan T, Stifani S. Characterization of Human iPSC-derived Spinal Motor Neurons by Single-cell RNA Sequencing. Neuroscience 2020; 450:57-70. [PMID: 32380268 DOI: 10.1016/j.neuroscience.2020.04.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 01/09/2023]
Abstract
Human induced pluripotent stem cells (iPSCs) offer the opportunity to generate specific cell types from healthy and diseased individuals, allowing the study of mechanisms of early human development, modelling a variety of human diseases, and facilitating the development of new therapeutics. Human iPSC-based applications are often limited by the variability among iPSC lines originating from a single donor, as well as the heterogeneity among specific cell types that can be derived from iPSCs. The ability to deeply phenotype different iPSC-derived cell types is therefore of primary importance to the successful and informative application of this technology. Here we describe a combination of motor neuron (MN) derivation and single-cell RNA sequencing approaches to generate and characterize specific MN subtypes obtained from human iPSCs. Our studies provide evidence for rapid and robust generation of MN progenitor cells that can give rise to a heterogenous population of MNs. Approximately 58% of human iPSC-derived MNs display molecular characteristics of lateral motor column MNs, with a number of molecularly distinct subpopulations present within this MN group. Roughly 19% of induced MNs resemble hypaxial motor column MNs, while ∼6% of induced MNs have features of median motor column MNs. The present study has the potential to improve our understanding of iPSC-derived MN subtype function and dysfunction, possibly leading to improved iPSC-based applications for the study of human MN biology and disease.
Collapse
Affiliation(s)
- Louise Thiry
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801, rue University, Montreal (Quebec) H3A 2B4, Canada
| | - Regan Hamel
- Department of Clinical Neurosciences, Clifford Allbutt Building - Cambridge Biosciences Campus and NIHR Biomedical Research Centre, University of Cambridge, Hills Road, CB2 0HA Cambridge, UK
| | - Stefano Pluchino
- Department of Clinical Neurosciences, Clifford Allbutt Building - Cambridge Biosciences Campus and NIHR Biomedical Research Centre, University of Cambridge, Hills Road, CB2 0HA Cambridge, UK
| | - Thomas Durcan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801, rue University, Montreal (Quebec) H3A 2B4, Canada; Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, Canada
| | - Stefano Stifani
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801, rue University, Montreal (Quebec) H3A 2B4, Canada.
| |
Collapse
|
9
|
Drp1-Zip1 Interaction Regulates Mitochondrial Quality Surveillance System. Mol Cell 2018; 73:364-376.e8. [PMID: 30581142 DOI: 10.1016/j.molcel.2018.11.009] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 09/11/2018] [Accepted: 11/07/2018] [Indexed: 01/09/2023]
Abstract
Mitophagy, a mitochondrial quality control process for eliminating dysfunctional mitochondria, can be induced by a response of dynamin-related protein 1 (Drp1) to a reduction in mitochondrial membrane potential (MMP) and mitochondrial division. However, the coordination between MMP and mitochondrial division for selecting the damaged portion of the mitochondrial network is less understood. Here, we found that MMP is reduced focally at a fission site by the Drp1 recruitment, which is initiated by the interaction of Drp1 with mitochondrial zinc transporter Zip1 and Zn2+ entry through the Zip1-MCU complex. After division, healthy mitochondria restore MMP levels and participate in the fusion-fission cycle again, but mitochondria that fail to restore MMP undergo mitophagy. Thus, interfering with the interaction between Drp1 and Zip1 blocks the reduction of MMP and the subsequent mitophagic selection of damaged mitochondria. These results suggest that Drp1-dependent fission provides selective pressure for eliminating "bad sectors" in the mitochondrial network, serving as a mitochondrial quality surveillance system.
Collapse
|
10
|
Kosykh A, Beilin A, Sukhinich K, Vorotelyak E. Postnatal neural crest stem cells from hair follicle interact with nerve tissue in vitro and in vivo. Tissue Cell 2018; 54:94-104. [PMID: 30309515 DOI: 10.1016/j.tice.2018.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 01/05/2023]
Abstract
Neural crest stem cells that located in the postnatal hair follicle (HF-NCSC) are considered a promising tool for treatment of nervous system diseases and injuries. It is well known that HF-NCSC can be used in the spinal cord and sciatic nerve reparation but their ability to restore brain structures is poorly studied. In this article we are investigating the interaction between HF-NCSC and a nerve tissue (embryonic and adult). We have found out that HF-NCSC isolated from adult mice grow and differentiate in accordance with the mouse embryo developmental stage when co-cultured with the embryonic nerve tissue. The HF-NCSC migration is slower in the late embryonic tissue co-culture system compared to the early one. This phenomenon is related to the motor function of the cells but not to their proliferation level. We have demonstrated that the embryonic nerve tissue maintains HF-NCSC an undifferentiated status, while an adult brain tissue inhibits the cell proliferation and activates the differentiation processes. Besides, HF-NCSC pre-differentiated into the neuronal direction shows a higher survival and migration rate after the transplantation into the adult brain tissue compared to the undifferentiated HF-NCSC. Thus, we have investigated the postnatal HF-NCSC response to the nerve tissue microenvironment to analyze their possible application to the brain repair processes.
Collapse
Affiliation(s)
- Anastasiia Kosykh
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Vavilova 26, 119334, Moscow, Russian Federation; Pirogov Russian National Research Medical University, Ostrovitianova 1, 117997, Moscow, Russian Federation.
| | - Arkadii Beilin
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Vavilova 26, 119334, Moscow, Russian Federation
| | - Kirill Sukhinich
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Vavilova 26, 119334, Moscow, Russian Federation
| | - Ekaterina Vorotelyak
- Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Vavilova 26, 119334, Moscow, Russian Federation; Pirogov Russian National Research Medical University, Ostrovitianova 1, 117997, Moscow, Russian Federation; Lomonosov Moscow State University, Leninskie Gory 1, Moscow, Russian Federation
| |
Collapse
|
11
|
Pernet V. Nogo-A in the visual system development and in ocular diseases. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1300-1311. [PMID: 28408340 DOI: 10.1016/j.bbadis.2017.04.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/08/2017] [Accepted: 04/09/2017] [Indexed: 01/02/2023]
Abstract
Nogo-A is a potent myelin-associated inhibitor for neuronal growth and plasticity in the central nervous system (CNS). Its effects are mediated by the activation of specific receptors that intracellularly control cytoskeleton rearrangements, protein synthesis and gene expression. Moreover, Nogo-A has been involved in the development of the visual system and in a variety of neurodegenerative diseases and injury processes that can alter its function. For example, Nogo-A was shown to influence optic nerve myelinogenesis, the formation and maturation of retinal axon projections, and retinal angiogenesis. In adult animals, the inactivation of Nogo-A exerted remarkable effects on visual plasticity. Relieving Nogo-A-induced inhibition increased axonal sprouting after optic nerve lesion and axonal rewiring in the visual cortex of intact adult mice. This review aims at presenting our current knowledge on the role of Nogo-A in the visual system and to discuss how its therapeutic targeting may promote visual improvement in ophthalmic diseases.
Collapse
Affiliation(s)
- Vincent Pernet
- CUO-Recherche, Centre de recherche du CHU de Québec and Département d'ophtalmologie, Faculté de médecine, Université Laval, Québec, Québec, Canada.
| |
Collapse
|
12
|
Garcia TB, Hollborn M, Bringmann A. Expression and signaling of NGF in the healthy and injured retina. Cytokine Growth Factor Rev 2017; 34:43-57. [PMID: 27964967 DOI: 10.1016/j.cytogfr.2016.11.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 11/28/2016] [Indexed: 01/02/2023]
Abstract
This review summarizes the present knowledge concerning the retinal localization of the nerve growth factor (NGF), its precursor proNGF, and the receptors TrkA and p75NTR in the developing and mature rodent retina. We further discuss the changes in the expression of NGF and the receptors in experimental models of retinal disorders and diseases like inherited retinitis pigmentosa, retinal detachment, glaucoma, and diabetic retinopathy. Since proNGF is now recognized as a bioactive signaling molecule which induces cell death through p75NTR activation, the role of proNGF in the induction of retinal cell loss under neurodegenerative conditions is also highlighted. In addition, we present the evidences for a potential therapeutic intervention with NGF for the treatment of retinal neurodegenerative diseases. Different strategies have been developed and experimentally tested in mice and rats in order to reduce cell loss and Müller cell gliosis, e.g., increasing the availability of endogenous NGF, administration of exogenous NGF, activation of TrkA, and inhibition of p75NTR. Here, we discuss the several lines of evidence supporting a protective effect of NGF on retinal cell loss, with specific emphasis on photoreceptor and retinal ganglion cell degeneration. A better understanding of the mechanisms underlying the effects of NGF and proNGF in the modulation of neurodegeneration and gliosis in the retina will help to develop efficient therapeutic strategies for various retinal diseases.
Collapse
Affiliation(s)
| | - Margrit Hollborn
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| | - Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| |
Collapse
|
13
|
Retrograde apoptotic signaling by the p75 neurotrophin receptor. Neuronal Signal 2017; 1:NS20160007. [PMID: 32714573 PMCID: PMC7373242 DOI: 10.1042/ns20160007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 02/06/2023] Open
Abstract
Neurotrophins are target-derived factors necessary for mammalian nervous system development and maintenance. They are typically produced by neuronal target tissues and interact with their receptors at axonal endings. Therefore, locally generated neurotrophin signals must be conveyed from the axon back to the cell soma. Retrograde survival signaling by neurotrophin binding to Trk receptors has been extensively studied. However, neurotrophins also bind to the p75 receptor, which can induce apoptosis in a variety of contexts. Selective activation of p75 at distal axon ends has been shown to generate a retrograde apoptotic signal, although the mechanisms involved are poorly understood. The present review summarizes the available evidence for retrograde proapoptotic signaling in general and the role of the p75 receptor in particular, with discussion of unanswered questions in the field. In-depth knowledge of the mechanisms of retrograde apoptotic signaling is essential for understanding the etiology of neurodegeneration in many diseases and injuries.
Collapse
|
14
|
Elshaer SL, El-Remessy AB. Implication of the neurotrophin receptor p75 NTR in vascular diseases: beyond the eye. EXPERT REVIEW OF OPHTHALMOLOGY 2016; 12:149-158. [PMID: 28979360 DOI: 10.1080/17469899.2017.1269602] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION The p75 neurotrophin receptor (p75NTR) is a member of TNF-α receptor superfamily that bind all neurotrophins, mainly regulating their pro-apoptotic actions. Ischemia is a common pathology in different cardiovascular diseases affecting multiple organs, however the contribution of p75NTR remains not fully addressed. The aim of this work is to review the current evidence through published literature studying the impact of p75NTR receptor in ischemic vascular diseases. AREAS COVERED In the eye, several ischemic ocular diseases are associated with enhanced p75NTR expression. Ischemic retinopathy including diabetic retinopathy, retinopathy of prematurity and retinal vein occlusion are characterized initially by ischemia followed by excessive neovascularization. Beyond the eye, cerebral ischemia, myocardial infarction and critical limb ischemia are ischemic cardiovascular diseases that are characterized by altered expression of neurotrophins and p75NTR expression. We surveyed both clinical and experimental studies that examined the impact of p75NTR receptor in ischemic diseases of eye, heart, brain and peripheral limbs. EXPERT COMMENTARY p75NTR receptor is a major player in multiple ischemic vascular diseases affecting the eye, brain, heart and peripheral limbs with significant increases in its expression accompanying neuro-vascular injury. This has been addressed in the current review along with the beneficial vascular outcomes of p75NTR inhibition.
Collapse
Affiliation(s)
- Sally L Elshaer
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, USA.,Research Service, Charlie Norwood VA Medical Center, Augusta, GA
| | - Azza B El-Remessy
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, USA.,Research Service, Charlie Norwood VA Medical Center, Augusta, GA.,Augusta Biomedical Research Corporation, Augusta, GA, USA
| |
Collapse
|
15
|
Abstract
Neurons are usually regarded as postmitotic cells that undergo apoptosis in response to cell cycle reactivation. Nevertheless, recent evidence indicates the existence of a defined developmental program that induces DNA replication in specific populations of neurons, which remain in a tetraploid state for the rest of their adult life. Similarly, de novo neuronal tetraploidization has also been described in the adult brain as an early hallmark of neurodegeneration. The aim of this review is to integrate these recent developments in the context of cell cycle regulation and apoptotic cell death in neurons. We conclude that a variety of mechanisms exists in neuronal cells for G1/S and G2/M checkpoint regulation. These mechanisms, which are connected with the apoptotic machinery, can be modulated by environmental signals and the neuronal phenotype itself, thus resulting in a variety of outcomes ranging from cell death at the G1/S checkpoint to full proliferation of differentiated neurons.
Collapse
Key Words
- AD, Alzheimer disease
- BDNF, brain-derived neurotrophic factor
- BrdU, 5-bromo-2′-deoxyuridine
- CKI, Cdk-inhibitor
- CNS, central nervous system
- Cdk, cyclin-dependent kinase
- Cip/Kip, cyclin inhibitor protein/kinase inhibitor protein
- G0, quiescent state
- G1, growth phase 1
- G2, growth phase 2
- Ink, inhibitor of kinase
- Mcm2, minichromosome maintenance 2
- PCNA, proliferating cell nuclear antigen
- PD, Parkinson disease
- RGCs, retinal ganglion cells
- Rb, Retinoblastoma
- S-phase
- S-phase, synthesis phase.
- apoptosis
- cell cycle re-entry
- mitosis
- neuron
- p38MAPK, p38 mitogen-activated protein kinase
- p75NTR, neurotrophin receptor p75
- tetraploid
Collapse
Affiliation(s)
- José M Frade
- a Department of Molecular, Cellular and Developmental Neurobiology; Instituto Cajal; Consejo Superior de Investigaciones Científicas (IC-CSIC) ; Madrid , Spain
| | | |
Collapse
|
16
|
Kraemer BR, Snow JP, Vollbrecht P, Pathak A, Valentine WM, Deutch AY, Carter BD. A role for the p75 neurotrophin receptor in axonal degeneration and apoptosis induced by oxidative stress. J Biol Chem 2014; 289:21205-16. [PMID: 24939843 PMCID: PMC4118083 DOI: 10.1074/jbc.m114.563403] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 06/10/2014] [Indexed: 12/14/2022] Open
Abstract
The p75 neurotrophin receptor (p75(NTR)) mediates the death of specific populations of neurons during the development of the nervous system or after cellular injury. The receptor has also been implicated as a contributor to neurodegeneration caused by numerous pathological conditions. Because many of these conditions are associated with increases in reactive oxygen species, we investigated whether p75(NTR) has a role in neurodegeneration in response to oxidative stress. Here we demonstrate that p75(NTR) signaling is activated by 4-hydroxynonenal (HNE), a lipid peroxidation product generated naturally during oxidative stress. Exposure of sympathetic neurons to HNE resulted in neurite degeneration and apoptosis. However, these effects were reduced markedly in neurons from p75(NTR-/-) mice. The neurodegenerative effects of HNE were not associated with production of neurotrophins and were unaffected by pretreatment with a receptor-blocking antibody, suggesting that oxidative stress activates p75(NTR) via a ligand-independent mechanism. Previous studies have established that proteolysis of p75(NTR) by the metalloprotease TNFα-converting enzyme and γ-secretase is necessary for p75(NTR)-mediated apoptotic signaling. Exposure of sympathetic neurons to HNE resulted in metalloprotease- and γ-secretase-dependent cleavage of p75(NTR). Pharmacological blockade of p75(NTR) proteolysis protected sympathetic neurons from HNE-induced neurite degeneration and apoptosis, suggesting that cleavage of p75(NTR) is necessary for oxidant-induced neurodegeneration. In vivo, p75(NTR-/-) mice exhibited resistance to axonal degeneration associated with oxidative injury following administration of the neurotoxin 6-hydroxydopamine. Together, these data suggest a novel mechanism linking oxidative stress to ligand-independent cleavage of p75(NTR), resulting in axonal fragmentation and neuronal death.
Collapse
Affiliation(s)
| | | | | | | | - William M Valentine
- Pathology, Microbiology, and Immunology, the Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN 37232
| | | | | |
Collapse
|
17
|
Jia YX, Li JR, Mao CY, Yin WT, Jiang RH. Glycyrrhizin improves p75NTR-associated sciatic nerve regeneration in a BALB/c mouse model. Exp Ther Med 2014; 7:1141-1146. [PMID: 24940400 PMCID: PMC3991491 DOI: 10.3892/etm.2014.1546] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 02/10/2014] [Indexed: 12/22/2022] Open
Abstract
Glycyrrhizin has a role in immune regulation in the central nervous system, but its impact on sciatic nerve injury had not previously been reported. In this study, a BALB/c mouse model of sciatic nerve injury was used to explore the role of glycyrrhizin in sciatic nerve repair and its underlying mechanism. Glycyrrhizin with intragastric gavage of 10 and 20 mg/kg weight per day (mid- and high-dose, respectively) inhibited p75 neurotrophin receptor (p75NTR) expression at the protein and mRNA levels versus the 5 mg/kg (low-dose) group and control (0.9% NaCl solution) at one, two, four and eight weeks following sciatic nerve injury, and simultaneously improved the action potential amplitude and motor nerve conductive velocity. Combined Marsland, Glees and Erikson’s silver stain and Luxol fast blue staining results indicated that high- and mid-dose glycyrrhizin promoted improved sciatic nerve myelination compared with the low-dose or control groups eight weeks after injury. Immunofluorescence staining demonstrated that glycyrrhizin had an inhibitory effect to a certain degree on local hypertrophic scar and inflammatory responses in the mouse model. In conclusion, glycyrrhizin can promote sciatic nerve regeneration and functional repair, in which doses of 10 and 20 mg/kg per day are more effective than lower doses, and such regeneration is associated with the downregulation of p75NTR.
Collapse
Affiliation(s)
- Yu-Xi Jia
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Jin-Ran Li
- Jilin University Bethune School of Medical Sciences, Changchun, Jilin 130021, P.R. China
| | - Cui-Ying Mao
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Wei-Tian Yin
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Ri-Hua Jiang
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
18
|
Andersen JL, Schrøder TJ, Christensen S, Strandbygård D, Pallesen LT, García-Alai MM, Lindberg S, Langgård M, Eskildsen JC, David L, Tagmose L, Simonsen KB, Maltas PJ, Rønn LCB, de Jong IEM, Malik IJ, Egebjerg J, Karlsson JJ, Uppalanchi S, Sakumudi DR, Eradi P, Watson SP, Thirup S. Identification of the first small-molecule ligand of the neuronal receptor sortilin and structure determination of the receptor-ligand complex. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2014; 70:451-60. [PMID: 24531479 PMCID: PMC3940197 DOI: 10.1107/s1399004713030149] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 11/04/2013] [Indexed: 01/03/2023]
Abstract
Sortilin is a type I membrane glycoprotein belonging to the vacuolar protein sorting 10 protein (Vps10p) family of sorting receptors and is most abundantly expressed in the central nervous system. Sortilin has emerged as a key player in the regulation of neuronal viability and has been implicated as a possible therapeutic target in a range of disorders. Here, the identification of AF40431, the first reported small-molecule ligand of sortilin, is reported. Crystals of the sortilin-AF40431 complex were obtained by co-crystallization and the structure of the complex was solved to 2.7 Å resolution. AF40431 is bound in the neurotensin-binding site of sortilin, with the leucine moiety of AF40431 mimicking the binding mode of the C-terminal leucine of neurotensin and the 4-methylumbelliferone moiety of AF40431 forming π-stacking with a phenylalanine.
Collapse
Affiliation(s)
- Jacob Lauwring Andersen
- The Lundbeck Foundation Research Centre MIND, Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus C, Denmark
| | - Tenna Juul Schrøder
- Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Søren Christensen
- Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Dorthe Strandbygård
- The Lundbeck Foundation Research Centre MIND, Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus C, Denmark
| | - Lone Tjener Pallesen
- The Lundbeck Foundation Research Centre MIND, Department of Biomedicine, Aarhus University, Ole Worms Allé 3, 8000 Aarhus C, Denmark
| | - Maria Marta García-Alai
- The Lundbeck Foundation Research Centre MIND, Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus C, Denmark
| | - Samsa Lindberg
- Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Morten Langgård
- Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | | | - Laurent David
- Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Lena Tagmose
- Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Klaus Baek Simonsen
- Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Philip James Maltas
- Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | | | - Inge E. M. de Jong
- Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Ibrahim John Malik
- Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Jan Egebjerg
- Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Jens-Jacob Karlsson
- Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Srinivas Uppalanchi
- Medicinal Chemistry, GVK BioScience, Plot No. 28 A, IDA Nacharam, Hyderabad 500 076, India
| | - Durga Rao Sakumudi
- Medicinal Chemistry, GVK BioScience, Plot No. 28 A, IDA Nacharam, Hyderabad 500 076, India
| | - Pradheep Eradi
- Medicinal Chemistry, GVK BioScience, Plot No. 28 A, IDA Nacharam, Hyderabad 500 076, India
| | - Steven P. Watson
- Neuroscience Drug Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Søren Thirup
- The Lundbeck Foundation Research Centre MIND, Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus C, Denmark
| |
Collapse
|
19
|
Kraemer BR, Yoon SO, Carter BD. The biological functions and signaling mechanisms of the p75 neurotrophin receptor. Handb Exp Pharmacol 2014; 220:121-164. [PMID: 24668472 DOI: 10.1007/978-3-642-45106-5_6] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The p75 neurotrophin receptor (p75(NTR)) regulates a wide range of cellular functions, including programmed cell death, axonal growth and degeneration, cell proliferation, myelination, and synaptic plasticity. The multiplicity of cellular functions governed by the receptor arises from the variety of ligands and co-receptors which associate with p75(NTR) and regulate its signaling. P75(NTR) promotes survival through interactions with Trk receptors, inhibits axonal regeneration via partnerships with Nogo receptor (Nogo-R) and Lingo-1, and promotes apoptosis through association with Sortilin. Signals downstream of these interactions are further modulated through regulated intramembrane proteolysis (RIP) of p75(NTR) and by interactions with numerous cytosolic partners. In this chapter, we discuss the intricate signaling mechanisms of p75(NTR), emphasizing how these signals are differentially regulated to mediate these diverse cellular functions.
Collapse
Affiliation(s)
- B R Kraemer
- Department of Biochemistry, Vanderbilt University School of Medicine, 625 Light Hall, Nashville, TN, 37232, USA
| | | | | |
Collapse
|
20
|
Abstract
Like most growth factors, neurotrophins are initially synthesized as precursors that are cleaved to release C-terminal mature forms. The well-characterized mature neurotrophins bind to Trk receptors to initiate survival and differentiative responses. More recently, the precursor forms or proneurotrophins have been found to act as distinct ligands by binding to an unrelated receptor complex consisting of the p75 neurotrophin receptor (p75) and sortilin to initiate cell death. Induction of proNGF and p75 has been observed in preclinical injury models and in pathological states in the central nervous system, and strategies that block the proNGF/p75 interaction are effective in limiting neuronal apoptosis. In contrast, the mechanisms that regulate expression of other proneurotrophins, including proBDNF and proNT-3, are less well understood. Here, recent findings on the biological actions, regulation of expression, and pathophysiological effects of proneurotrophins will be reviewed.
Collapse
Affiliation(s)
- B L Hempstead
- Department of Medicine, Weill Cornell Medical College, Room C610, 1300 York Ave, New York, NY, 10065, USA,
| |
Collapse
|
21
|
Ceni C, Unsain N, Zeinieh MP, Barker PA. Neurotrophins in the regulation of cellular survival and death. Handb Exp Pharmacol 2014; 220:193-221. [PMID: 24668474 DOI: 10.1007/978-3-642-45106-5_8] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The neurotrophins play crucial roles regulating survival and apoptosis in the developing and injured nervous system. The four neurotrophins exert profound and crucial survival effects on developing peripheral neurons, and their expression and action is intimately tied to successful innervation of peripheral targets. In the central nervous system, they are dispensable for neuronal survival during development but support neuronal survival after lesion or other forms of injury. Neurotrophins also regulate apoptosis of both peripheral and central neurons, and we now recognize that there are regulatory advantages to having the same molecules regulate life and death decisions. This chapter examines the biological contexts in which these events take place and highlights the specific ligands, receptors, and signaling mechanisms that allow them to occur.
Collapse
Affiliation(s)
- Claire Ceni
- Centre for Neuronal Survival, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC, Canada, H3A 2B4
| | | | | | | |
Collapse
|
22
|
Braunger BM, Demmer C, Tamm ER. Programmed cell death during retinal development of the mouse eye. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 801:9-13. [PMID: 24664675 DOI: 10.1007/978-1-4614-3209-8_2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Similar to other parts of the central nervous system, there are two types of programmed cell death during retinal development. In early development, the neuronal progenitor population is affected. In the mouse eye, this kind of programmed cell death begins at around embryonic day (E) 12.5 and peaks between E14.5 and E16.5. The second phase of programmed cell death occurs during synaptogenesis within the first 2 postnatal weeks. Important signaling mechanisms that induce programmed cell death of retinal progenitors appear to involve nerve growth factor acting on the proapoptotic receptor to p75 neurotrophin receptor (p75(NTR)) and transforming growth factor-β.
Collapse
Affiliation(s)
- Barbara M Braunger
- Institute of Human Anatomy and Embryology, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany
| | | | | |
Collapse
|
23
|
Sunagar K, Fry BG, Jackson TNW, Casewell NR, Undheim EAB, Vidal N, Ali SA, King GF, Vasudevan K, Vasconcelos V, Antunes A. Molecular evolution of vertebrate neurotrophins: co-option of the highly conserved nerve growth factor gene into the advanced snake venom arsenalf. PLoS One 2013; 8:e81827. [PMID: 24312363 PMCID: PMC3843689 DOI: 10.1371/journal.pone.0081827] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 10/17/2013] [Indexed: 01/19/2023] Open
Abstract
Neurotrophins are a diverse class of structurally related proteins, essential for neuronal development, survival, plasticity and regeneration. They are characterized by major family members, such as the nerve growth factors (NGF), brain-derived neurotrophic factors (BDNF) and neurotrophin-3 (NT-3), which have been demonstrated here to lack coding sequence variations and follow the regime of negative selection, highlighting their extremely important conserved role in vertebrate homeostasis. However, in stark contrast, venom NGF secreted as part of the chemical arsenal of the venomous advanced snake family Elapidae (and to a lesser extent Viperidae) have characteristics consistent with the typical accelerated molecular evolution of venom components. This includes a rapid rate of diversification under the significant influence of positive-selection, with the majority of positively-selected sites found in the secreted β-polypeptide chain (74%) and on the molecular surface of the protein (92%), while the core structural and functional residues remain highly constrained. Such focal mutagenesis generates active residues on the toxin molecular surface, which are capable of interacting with novel biological targets in prey to induce a myriad of pharmacological effects. We propose that caenophidian NGFs could participate in prey-envenoming by causing a massive release of chemical mediators from mast cells to mount inflammatory reactions and increase vascular permeability, thereby aiding the spread of other toxins and/or by acting as proapoptotic factors. Despite their presence in reptilian venom having been known for over 60 years, this is the first evidence that venom-secreted NGF follows the molecular evolutionary pattern of other venom components, and thus likely participates in prey-envenomation.
Collapse
Affiliation(s)
- Kartik Sunagar
- CIMAR/CIIMAR, Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Porto, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Bryan Grieg Fry
- Venom Evolution Lab, School of Biological Sciences, The University of Queensland, St. Lucia, Queensland, Australia
- Institute for Molecular Bioscience, University of Queenland, St Lucia, Queensland, Australia
| | - Timothy N. W. Jackson
- Venom Evolution Lab, School of Biological Sciences, The University of Queensland, St. Lucia, Queensland, Australia
- Institute for Molecular Bioscience, University of Queenland, St Lucia, Queensland, Australia
| | - Nicholas R. Casewell
- Molecular Ecology and Evolution Group, School of Biological Sciences, Bangor University, Bangor, United Kingdom
- Alistair Reid Venom Research Unit, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Eivind A. B. Undheim
- Venom Evolution Lab, School of Biological Sciences, The University of Queensland, St. Lucia, Queensland, Australia
- Institute for Molecular Bioscience, University of Queenland, St Lucia, Queensland, Australia
| | - Nicolas Vidal
- Département Systématique et Evolution, Service de Systématique Moléculaire, UMR 7138, Muséum National d’Histoire Naturelle, Paris, France
| | - Syed A. Ali
- Venom Evolution Lab, School of Biological Sciences, The University of Queensland, St. Lucia, Queensland, Australia
- HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, Pakistan
| | - Glenn F. King
- Institute for Molecular Bioscience, University of Queenland, St Lucia, Queensland, Australia
| | | | - Vitor Vasconcelos
- CIMAR/CIIMAR, Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Porto, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Agostinho Antunes
- CIMAR/CIIMAR, Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Porto, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- * E-mail:
| |
Collapse
|
24
|
Ovejero-Benito MC, Frade JM. Brain-derived neurotrophic factor-dependent cdk1 inhibition prevents G2/M progression in differentiating tetraploid neurons. PLoS One 2013; 8:e64890. [PMID: 23741412 PMCID: PMC3669015 DOI: 10.1371/journal.pone.0064890] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 04/19/2013] [Indexed: 11/28/2022] Open
Abstract
Neurodegeneration is often associated with DNA synthesis in neurons, the latter usually remaining for a long time as tetraploid cells before dying by apoptosis. The molecular mechanism preventing G2/M transition in these neurons remains unknown, but it may be reminiscent of the mechanism that maintains tetraploid retinal ganglion cells (RGCs) in a G2-like state during normal development, thus preventing their death. Here we show that this latter process, known to depend on brain-derived neurotrophic factor (BDNF), requires the inhibition of cdk1 by TrkB. We demonstrate that a subpopulation of chick RGCs previously shown to become tetraploid co-expresses TrkB and cdk1 in vivo. By using an in vitro system that recapitulates differentiation and cell cycle re-entry of chick retinal neurons we show that BDNF, employed at concentrations specific for the TrkB receptor, reduces the expression of cdk1 in TrkB-positive, differentiating neurons. In this system, BDNF also inhibits the activity of both endogenous cdk1 and exogenously-expressed cdk1/cyclin B1 complex. This inhibition correlates with the phosphorylation of cdk1 at Tyr15, an effect that can be prevented with K252a, a tyrosine kinase inhibitor commonly used to prevent the activity of neurotrophins through their Trk receptors. The effect of BDNF on cdk1 activity is Tyr15-specific since BDNF cannot prevent the activity of a constitutively active form of cdk1 (Tyr15Phe) when expressed in differentiating retinal neurons. We also show that BDNF-dependent phosphorylation of cdk1 at Tyr15 could not be blocked with MK-1775, a Wee1-selective inhibitor, indicating that Tyr15 phosphorylation in cdk1 does not seem to occur through the canonical mechanism observed in proliferating cells. We conclude that the inhibition of both expression and activity of cdk1 through a BDNF-dependent mechanism contributes to the maintenance of tetraploid RGCs in a G2-like state.
Collapse
Affiliation(s)
- María C. Ovejero-Benito
- Department of Molecular, Cellular, and Developmental Neurobiology, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - José M. Frade
- Department of Molecular, Cellular, and Developmental Neurobiology, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- * E-mail:
| |
Collapse
|
25
|
Kotlyanskaya L, McLinden KA, Giniger E. Of proneurotrophins and their antineurotrophic effects. Sci Signal 2013; 6:pe6. [PMID: 23405011 DOI: 10.1126/scisignal.2003824] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurotrophins perform essential processes throughout neural development. They signal through Trk receptor proteins, typically in association with a "low affinity" p75(NTR) pan-neurotrophin co-receptor. Neurotrophins are synthesized as proproteins; the pro domains are removed proteolytically to yield the mature, presumably functional forms of the neurotrophins. Recent findings, however, have revealed a positive role for the proneurotrophins themselves. The proproteins bind with high affinity to the p75(NTR) pan-neurotrophin receptor in the absence of Trks to initiate a separate set of signaling cascades that actively oppose the effects of the mature growth factors. These experiments suggest that the balance between pro- and mature neurotrophin plays a critical role in tuning downstream signaling. This view changes the neurotrophin field substantially and also points to the broader idea that the potential activities of precursor proteins deserve a closer look.
Collapse
Affiliation(s)
- Lucy Kotlyanskaya
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20854, USA
| | | | | |
Collapse
|
26
|
Neuronal programmed cell death-1 ligand expression regulates retinal ganglion cell number in neonatal and adult mice. J Neuroophthalmol 2013; 32:227-37. [PMID: 22635166 DOI: 10.1097/wno.0b013e3182589589] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVES During mouse retina maturation, the final number of retinal ganglion cells (RGCs) is determined by highly regulated programmed cell death. Previous studies demonstrated that the immunoregulatory receptor programmed cell death-1 (PD-1) promotes developmental RGC death. To identify the functional signaling partner(s) for PD-1, we identified retinal expression of PD-1 ligands and examined the effect of PD-1 ligand expression on RGC number. We also explored the hypothesis that PD-1 signaling promotes the development of functional visual circuitry. METHODS Characterization of retinal and brain programmed cell death-1 ligand 1 (PD-L1) expression were examined by immunofluorescence on tissue sections. The contribution of PD-ligands, PD-L1, and programmed cell death-1 ligand 2 (PD-L2) to RGC number was examined in PD-ligand knockout mice lacking 1 or both ligands. Retinal architecture was assessed by spectral-domain optical coherence tomography, and retinal function was analyzed by electroretinography in wild-type and PD-L1/L2 double-deficient mice. RESULTS PD-L1 expression is found throughout the neonatal retina and persists in adult RGCs, bipolar interneurons, and Müller glia. In the absence of both PD-ligands, there is a significant numerical increase in RGCs (34% at postnatal day 2 [P2] and 18% in adult), as compared to wild type, and PD-ligands have redundant function in this process. Despite the increased RGC number, adult PD-L1/L2 double-knockout mice have normal retinal architecture and outer retina function. CONCLUSION This study demonstrates that PD-L1 and PD-L2 together impact the final number of RGCs in adult mice and supports a novel role for active promotion of neuronal cell death through PD-1 receptor-ligand engagement.
Collapse
|
27
|
Le Moan N, Houslay DM, Christian F, Houslay MD, Akassoglou K. Oxygen-dependent cleavage of the p75 neurotrophin receptor triggers stabilization of HIF-1α. Mol Cell 2011; 44:476-90. [PMID: 22055192 PMCID: PMC3212815 DOI: 10.1016/j.molcel.2011.08.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 05/23/2011] [Accepted: 08/15/2011] [Indexed: 12/23/2022]
Abstract
Homeostatic control of oxygen availability allows cells to survive oxygen deprivation. Although the transcription factor hypoxia-inducible factor 1α (HIF-1α) is the main regulator of the hypoxic response, the upstream mechanisms required for its stabilization remain elusive. Here, we show that p75 neurotrophin receptor (p75(NTR)) undergoes hypoxia-induced γ-secretase-dependent cleavage to provide a positive feed-forward mechanism required for oxygen-dependent HIF-1α stabilization. The intracellular domain of p75(NTR) directly interacts with the evolutionarily conserved zinc finger domains of the E3 RING ubiquitin ligase Siah2 (seven in absentia homolog 2), which regulates HIF-1α degradation. p75(NTR) stabilizes Siah2 by decreasing its auto-ubiquitination. Genetic loss of p75(NTR) dramatically decreases Siah2 abundance, HIF-1α stabilization, and induction of HIF-1α target genes in hypoxia. p75(NTR-/-) mice show reduced HIF-1α stabilization, vascular endothelial growth factor (VEGF) expression, and neoangiogenesis after retinal hypoxia. Thus, hypoxia-induced intramembrane proteolysis of p75(NTR) constitutes an apical oxygen-dependent mechanism to control the magnitude of the hypoxic response.
Collapse
Affiliation(s)
- Natacha Le Moan
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Daniel M. Houslay
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Frank Christian
- Molecular Pharmacology Group, Biochemistry & Molecular Biology, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | - Miles D. Houslay
- Molecular Pharmacology Group, Biochemistry & Molecular Biology, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | - Katerina Akassoglou
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
28
|
TRPV1 receptors modulate retinal development. Int J Dev Neurosci 2011; 29:405-13. [PMID: 21414401 DOI: 10.1016/j.ijdevneu.2011.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 01/19/2011] [Accepted: 03/08/2011] [Indexed: 12/11/2022] Open
Abstract
We investigated the possible participation of TRPV1 channels in retinal apoptosis and overall development. Retinas from newborn, male albino rats were treated in vitro with capsazepine, a TRPV1 antagonist. The expression of cell cycle markers was not changed after TRPV1 blockade, whereas capsazepine reduced the number of apoptotic cells throughout the retina,increased ERK1/2 and p38 phosphorylation and slightly reduced JNK phosphorylation. The expression of BAD, Bcl-2, as well as integral and cleaved capsase-3 were similar in all experimental conditions. Newborn rats were kept for 2 months after receiving high doses of capsazepine. In their retinas, calbindin and parvalbumin protein levels were upregulated, but only the number of amacrine-like, parvalbumin-positive cells was increased. The numbers of calretinin, calbindin, ChAT, vimentin, PKC-alpha and GABA-positive cells were similar in both conditions. Protein expression of synapsin Ib was also increased in the retinas of capsazepine-treated rats. Calretinin, vimentin, GFAP, synapsin Ia, synaptophysin and light neurofilament protein levels were not changed when compared to control values. Our results indicate that TRPV1 channels play a role in the control of the early apoptosis that occur during retinal development, which might be dependent on MAPK signaling. Moreover, it seems that TRPV1 function might be important for neuronal and synaptic maturation in the retina.
Collapse
|
29
|
Golbs A, Nimmervoll B, Sun JJ, Sava IE, Luhmann HJ. Control of programmed cell death by distinct electrical activity patterns. Cereb Cortex 2010; 21:1192-202. [PMID: 20966045 DOI: 10.1093/cercor/bhq200] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Electrical activity and sufficient supply with survival factors play a major role in the control of apoptosis in the developing cortex. Coherent high-frequency neuronal activity, which efficiently releases neurotrophins, is essential for the survival of immature neurons. We studied the influence of neuronal activity on apoptosis in the developing cortex. Dissociated cultures of the newborn mouse cerebral cortex were grown on multielectrode arrays to determine the activity patterns that promote neuronal survival. Cultures were transfected with a plasmid coding for a caspase-3-sensitive fluorescent protein allowing real-time analysis of caspase-3-dependent apoptosis in individual neurons. Elevated extracellular potassium concentrations (5 and 8 mM), application of 4-aminopyridine or the γ-aminobutyric acid-A receptor antagonist Gabazine induced a shift in the frequency distribution of activity toward high-frequency bursts. Under these conditions, a reduction or delay in caspase-3 activation and an overall increase in neuronal survival could be observed. This effect was dependent on the activity of phosphatidylinositol-3 kinase, as blockade of this enzyme abolished the survival-promoting effect of high extracellular potassium concentrations. Our data indicate that increased network activity can prevent apoptosis in developing cortical neurons.
Collapse
Affiliation(s)
- Antje Golbs
- Institute of Physiology and Pathophysiology, University Medical Center, Johannes Gutenberg University, D-55128 Mainz, Germany
| | | | | | | | | |
Collapse
|
30
|
Neurotrophin receptors TrkA and TrkC cause neuronal death whereas TrkB does not. Nature 2010; 467:59-63. [PMID: 20811452 DOI: 10.1038/nature09336] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Accepted: 07/06/2010] [Indexed: 11/08/2022]
Abstract
Neurons of the peripheral nervous system have long been known to require survival factors to prevent their death during development. But why they selectively become dependent on secretory molecules has remained a mystery, as is the observation that in the central nervous system, most neurons do not show this dependency. Using engineered embryonic stem cells, we show here that the neurotrophin receptors TrkA and TrkC (tropomyosin receptor kinase A and C, also known as Ntrk1 and Ntrk3, respectively) instruct developing neurons to die, both in vitro and in vivo. By contrast, TrkB (also known as Ntrk2), a closely related receptor primarily expressed in the central nervous system, does not. These results indicate that TrkA and TrkC behave as dependence receptors, explaining why developing sympathetic and sensory neurons become trophic-factor-dependent for survival. We suggest that the expansion of the Trk gene family that accompanied the segregation of the peripheral from the central nervous system generated a novel mechanism of cell number control.
Collapse
|
31
|
Brodski C, Vogt Weisenhorn DM, Dechant G. Therapy of neurodegenerative diseases using neurotrophic factors: cell biological perspective. Expert Rev Neurother 2010; 2:417-26. [PMID: 19810873 DOI: 10.1586/14737175.2.3.417] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurotrophic growth factors are a structurally diverse group of endogenous proteins, which profoundly regulate survival and multiple other cellular functions of healthy and diseased neurons. Their physiological targets include all neuronal cell types affected by neurodegenerative diseases. Initial clinical trials, which were based on strong and convincing neuroprotective effects in animal models of neurodegenerative diseases, have failed so far. In this review, we adopt a cell biological perspective to outline both the potential and problems of the therapeutic use of trophic factors in the nervous system. Emphasis is given to the functional interplay of neurotrophic factors in the regulation of cell death, as well as to the effects these proteins exert on neuronal morphology and synaptic transmission.
Collapse
Affiliation(s)
- Claude Brodski
- GSF Research Center, Clinical Neurogenetics, Ingolstädter, Landstrasse 1, Neuherberg, Germany.
| | | | | |
Collapse
|
32
|
Teng KK, Felice S, Kim T, Hempstead BL. Understanding proneurotrophin actions: Recent advances and challenges. Dev Neurobiol 2010; 70:350-9. [PMID: 20186707 DOI: 10.1002/dneu.20768] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurotrophins are initially synthesized as larger precursors (proneurotrophins), which undergo proteolytic cleavage to yield mature forms. Although the functions of the mature neurotrophins have been well established during neural development and in the adult nervous system, roles for the proneurotrophins in developmental and injury-induced cell death, as well as in synaptic plasticity, have only recently been appreciated. Interestingly, both mature neurotrophins and proneurotrophins utilize dual-receptor complexes to mediate their actions. The mature neurotrophin coreceptors consist of the Trk receptor tyrosine kinases and p75(NTR), wherein Trk transduces survival and differentiative signaling, and p75(NTR) modulates the affinity and selectivity of Trk activation. On the other hand, proneurotrophins engage p75(NTR) and the structurally distinct coreceptor sortilin, to initiate p75(NTR)-dependent signal transduction cascade. Although the specificity of mature neurotrophins vs. proneurotrophins actions is due in part to the formation of distinct coreceptor complexes, a number of recent studies highlight how different p75(NTR)-mediated cellular actions are modulated. Here, we review emerging evidence for a novel transmembrane mechanism for ligand-specific p75(NTR) activation and several mechanisms by which p75(NTR)-dependent apoptotic and nonapoptotic responses can be selective activated.
Collapse
Affiliation(s)
- Kenneth K Teng
- Department of Medicine, Weill Cornell Medical College, New York, New York 10065, USA
| | | | | | | |
Collapse
|
33
|
Oldreive CE, Doherty GH. Effects of tumour necrosis factor-alpha on developing cerebellar granule and Purkinje neurons in vitro. J Mol Neurosci 2010; 42:44-52. [PMID: 20419354 DOI: 10.1007/s12031-010-9370-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 04/07/2010] [Indexed: 11/27/2022]
Abstract
Tumour necrosis factor-alpha (TNF-alpha) has been widely implicated in both neurodevelopment and neurodegeneration, yet its effects on individual populations of cerebellar neurons as they develop have not been fully elucidated. Therefore, we established primary neuronal cultures of developing murine cerebellar Purkinje neurons and postnatal cerebellar granule cells to determine the consequences of TNF-alpha exposure for their survival. We discovered that TNF-alpha did not affect the viability of cerebellar granule neurons at any of the ages studied, even though TNF-alpha and its receptors, TNFR1 and TNFR2, are widely expressed in the postnatal cerebellum. In addition, TNF-alpha was neither able to ameliorate, nor enhance, cell death in cerebellar granule cells elicited by a variety of stimuli including homocysteine and alcohol exposure. In contrast, in cultures established at embryonic day 16, TNF-alpha enhanced the number of cerebellar Purkinje neurons in vitro but this effect was not observed in embryonic day 19 cultures. Thus, TNF-alpha has differential and highly specific effects on different populations of cerebellar neurons as they develop.
Collapse
Affiliation(s)
- Ceri E Oldreive
- School of Biology, University of St Andrews, West Burn Lane, St Andrews, Fife KY169TS, Scotland, UK
| | | |
Collapse
|
34
|
ProNGF induces TNFalpha-dependent death of retinal ganglion cells through a p75NTR non-cell-autonomous signaling pathway. Proc Natl Acad Sci U S A 2010; 107:3817-22. [PMID: 20133718 DOI: 10.1073/pnas.0909276107] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neurotrophin binding to the p75 neurotrophin receptor (p75(NTR)) activates neuronal apoptosis following adult central nervous system injury, but the underlying cellular mechanisms remain poorly defined. In this study, we show that the proform of nerve growth factor (proNGF) induces death of retinal ganglion cells in adult rodents via a p75(NTR)-dependent signaling mechanism. Expression of p75(NTR) in the adult retina is confined to Müller glial cells; therefore we tested the hypothesis that proNGF activates a non-cell-autonomous signaling pathway to induce retinal ganglion cell (RGC) death. Consistent with this, we show that proNGF induced robust expression of tumor necrosis factor alpha (TNFalpha) in Müller cells and that genetic or biochemical ablation of TNFalpha blocked proNGF-induced death of retinal neurons. Mice rendered null for p75(NTR), its coreceptor sortilin, or the adaptor protein NRAGE were defective in proNGF-induced glial TNFalpha production and did not undergo proNGF-induced retinal ganglion cell death. We conclude that proNGF activates a non-cell-autonomous signaling pathway that causes TNFalpha-dependent death of retinal neurons in vivo.
Collapse
|
35
|
Sampieri K, Mencarelli MA, Carmela Epistolato M, Toti P, Lazzi S, Bruttini M, Francesco SD, Longo I, Meloni I, Mari F, Acquaviva A, Hadjistilianou T, Renieri A, Ariani F. Genomic differences between retinoma and retinoblastoma. Acta Oncol 2009; 47:1483-92. [PMID: 18785023 DOI: 10.1080/02841860802342382] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Genomic copy number changes are involved in the multi-step process transforming normal retina in retinoblastoma after RB1 mutational events. Previous studies on retinoblastoma samples led to a multi-step model in which after two successive RB1 mutations, further genomic changes accompany malignancy: 1q32.1 gain is followed by 6p22 gain, that in turn is followed by 16q22 loss and 2p24.1 gain. Retinoma is a benign variant of retinoblastoma that was initially considered a tumor regression, but recent evidences suggest that it rather represents a pre-malignant lesion. Genetic studies on retinoma tissue have rarely been performed. MATERIALS AND METHODS We investigated by Real-Time qPCR, copy number changes of candidate genes located within the 4 hot-spot regions (MDM4 at 1q32.1, MYCN at 2p24.1, E2F3 at 6p22 and CDH11 at 16q22) in retina, retinoma and retinoblastoma tissues from two different patients. RESULTS Our results demonstrated that some copy number changes thought to belong to early (MDM4 gain) or late stage (MYCN and E2F3 gain) of retinoblastoma are already present in retinoma at the same (for MDM4) or at lower (for MYCN and E2F3) copy number variation respect to retinoblastoma. CDH11 copy number is not altered in the two retinoma samples, but gain is present in one of the two retinoblastomas. DISCUSSION Our results suggest that MDM4 gain may be involved in the early transition from normal retina to retinoma, while MYCN and E2F3 progressive gain may represent driving factors of tumor progression. These results also confirm the pre-malignant nature of retinoma.
Collapse
|
36
|
Kim T, Hempstead BL. NRH2 is a trafficking switch to regulate sortilin localization and permit proneurotrophin-induced cell death. EMBO J 2009; 28:1612-23. [PMID: 19407813 DOI: 10.1038/emboj.2009.118] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Accepted: 03/30/2009] [Indexed: 12/25/2022] Open
Abstract
Proneurotrophins mediate neuronal apoptosis using a dual receptor complex of sortilin and p75(NTR). Although p75(NTR) is highly expressed on the plasma membrane and accessible to proneurotrophin ligands, sortilin is primarily localized to intracellular membranes, limiting the formation of a cell surface co-receptor complex. Here, we show that the mammalian p75(NTR) homologue NRH2 critically regulates the expression of sortilin on the neuronal cell surface and promotes p75(NTR) and sortilin receptor complex formation, rendering cells responsive to proneurotrophins. This is accomplished by interactions between the cytoplasmic domains of NRH2 and sortilin that impair lysosomal degradation of sortilin. In proneurotrophin-responsive neurons, acute silencing of endogenous NRH2 significantly reduces cell surface-expressed sortilin and abolishes proneurotrophin-induced neuronal death. Thus, these data suggest that NRH2 acts as a trafficking switch to impair lysosomal-dependant sortilin degradation and to redistribute sortilin to the cell surface, rendering p75(NTR)-expressing cells susceptible to proneurotrophin-induced death.
Collapse
Affiliation(s)
- Taeho Kim
- Graduate Program in Neuroscience, Weill Medical College of Cornell University, New York, NY 10065, USA
| | | |
Collapse
|
37
|
Hempstead BL. Commentary: Regulating proNGF action: multiple targets for therapeutic intervention. Neurotox Res 2009; 16:255-60. [PMID: 19526280 DOI: 10.1007/s12640-009-9054-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 03/23/2009] [Accepted: 04/03/2009] [Indexed: 01/26/2023]
Abstract
Neurotrophins are initially synthesized as precursor forms that are cleaved to release C-terminal mature forms that bind to Trk receptors to initiate survival and differentiative responses. Recent studies suggest that the precursor form of NGF (proNGF) acts as a distinct ligand by binding to a receptor complex of p75 and sortilin to initiate cell death. Induction of proNGF and p75 has been observed in multiple pathological states and injury models in the central nervous system, and blockade of proNGF/p75 interaction is efficacious in limiting neuronal apoptosis. Multiple strategies that may act to limit proNGF action are considered as potential therapeutic targets for future development.
Collapse
Affiliation(s)
- Barbara L Hempstead
- Department of Medicine, Weill Cornell Medical College, 1300 York Ave, New York, NY 10065, USA.
| |
Collapse
|
38
|
Catts VS, Al-Menhali N, Burne THJ, Colditz MJ, Coulson EJ. The p75 neurotrophin receptor regulates hippocampal neurogenesis and related behaviours. Eur J Neurosci 2008; 28:883-92. [PMID: 18717734 DOI: 10.1111/j.1460-9568.2008.06390.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Although changes to neural circuitry are believed to underlie behavioural characteristics mediated by the hippocampus, the contribution of neurogenesis to this process remains controversial. This is partially because the molecular regulators of neurogenesis remain to be fully elucidated, and experiments generically preventing neurogenesis have, for the most part, depended on paradigms involving irradiation. Here we show that mice lacking the p75 neurotrophin receptor (p75(NTR-/-)) have 25% fewer neuroblasts and 50% fewer newborn neurons in the dentate gyrus, coincident with increased rates of cell death of newly born cells and a significantly smaller granular cell layer and dentate gyrus, than those of p75(NTR+/+) mice. Whereas p75(NTR-/-) mice had increased latency to feed in a novelty-suppressed feeding paradigm they had increased mobility in another test of "depression", the tail-suspension test. p75(NTR-/-) mice also had subtle behavioural impairment in Morris water maze tasks compared to wild-type animals. No difference between genotypes was found in relation to anxiety or exploration behaviour based on the elevated-plus maze, light-dark, hole-board, T-maze or forced-swim tests. Overall, this study demonstrates that p75(NTR) is an important regulator of hippocampal neurogenesis, with concomitant effects on associated behaviours. However, the behavioural attributes of the p75(NTR-/-) mice may be better explained by altered circuitry driven by the loss of p75(NTR) in the basal forebrain, rather than direct changes to neurogenesis.
Collapse
Affiliation(s)
- Vibeke S Catts
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | | | | | | | | |
Collapse
|
39
|
de Melo Reis RA, Cabral-da-Silva MEC, de Mello FG, Taylor JSH. Müller glia factors induce survival and neuritogenesis of peripheral and central neurons. Brain Res 2008; 1205:1-11. [PMID: 18353289 DOI: 10.1016/j.brainres.2008.02.035] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2007] [Revised: 11/05/2007] [Accepted: 02/08/2008] [Indexed: 11/29/2022]
Abstract
We have examined the trophic effects of conditioned media obtained from purified murine Müller glia cells on chick purified sympathetic or dorsal root ganglia (DRG) neurons and on Retinal Ganglion Cells (RGC) from postnatal mice. Purified murine Müller glia cultures stained positively for vimentin, GFAP or S-100, but were negative for neuronal markers. Murine Müller glial conditioned medium (MMG) was concentrated and at 1:1 dilution supported 100% survival of chick or rat sympathetic neurons after 48 h compared to <5% in controls. Partial purification of the MMG using centriprep concentrators showed that trophic activity is from molecules above 10 kDa. MMG stimulated AKT, ERK and pStat3 in sympathetic neurons. Sympathetic or DRG neuronal survival induced by MMG was blocked by anti-human NGF, but not by anti-human CNTF (sympathetic) or by anti-BDNF (DRGs) neutralizing antibodies. MMG also induced neurite outgrowth in P4 mice retinal explants and on isolated RGC. RGCs plated on top of Müller glia cells had a much better survival rate (>80%, 96 h) compared to laminin+poly-L-lysine substrates. In conclusion, we show that purified mice Müller glia cultures secrete NGF that support peripheral neuronal survival and other unidentified trophic molecules that induce RGC survival and neuritogenesis.
Collapse
|
40
|
Dimaras H, Gallie BL. The p75NTR neurotrophin receptor is a tumor suppressor in human and murine retinoblastoma development. Int J Cancer 2008; 122:2023-9. [DOI: 10.1002/ijc.23356] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
41
|
de Melo J, Zhou QP, Zhang Q, Zhang S, Fonseca M, Wigle JT, Eisenstat DD. Dlx2 homeobox gene transcriptional regulation of Trkb neurotrophin receptor expression during mouse retinal development. Nucleic Acids Res 2007; 36:872-84. [PMID: 18086710 PMCID: PMC2241891 DOI: 10.1093/nar/gkm1099] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Dlx homeobox genes are first expressed in embryonic retina at E11.5. The Dlx1/Dlx2 null retina has a reduced ganglion cell layer (GCL), with loss of late-born differentiated retinal ganglion cells (RGCs) due to increased apoptosis. TrkB signaling is proposed to regulate the dynamics of RGC apoptosis throughout development. DLX2 expression markedly precedes the onset of TrkB expression in the GCL; TrkB co-expression with Dlx2 and RGC markers is well-established by E13.5. In the Dlx1/Dlx2 null retina, TrkB expression is significantly reduced by E16.5. We demonstrated that DLX2 binds to a specific region of the TrkB promoter in retinal neuroepithelium during embryogenesis. In vitro confirmation and the functional consequences of DLX2 binding to this TrkB regulatory region support TrkB as a Dlx2 transcriptional target. Furthermore, ectopic Dlx2 expression in retinal explants activates TrkB expression and Dlx2 knockdown in primary retinal cultures results in reduced TrkB expression. RGC differentiation and survival require the coordinated expression of transcription factors. This study establishes a direct transcriptional relationship between a homeodomain protein involved in RGC differentiation and a neurotrophin receptor implicated in RGC survival. Signaling mediated by TrkB may contribute to survival of late-born RGCs whose terminal differentiation is regulated by Dlx gene function.
Collapse
Affiliation(s)
- Jimmy de Melo
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | | | | | | |
Collapse
|
42
|
Jansen P, Giehl K, Nyengaard JR, Teng K, Lioubinski O, Sjoegaard SS, Breiderhoff T, Gotthardt M, Lin F, Eilers A, Petersen CM, Lewin GR, Hempstead BL, Willnow TE, Nykjaer A. Roles for the pro-neurotrophin receptor sortilin in neuronal development, aging and brain injury. Nat Neurosci 2007; 10:1449-57. [PMID: 17934455 DOI: 10.1038/nn2000] [Citation(s) in RCA: 214] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Accepted: 09/24/2007] [Indexed: 01/16/2023]
Abstract
Neurotrophins are essential for development and maintenance of the vertebrate nervous system. Paradoxically, although mature neurotrophins promote neuronal survival by binding to tropomyosin receptor kinases and p75 neurotrophin receptor (p75(NTR)), pro-neurotrophins induce apoptosis in cultured neurons by engaging sortilin and p75(NTR) in a death-signaling receptor complex. Substantial amounts of neurotrophins are secreted in pro-form in vivo, yet their physiological significance remains unclear. We generated a sortilin-deficient mouse to examine the contribution of the p75(NTR)/sortilin receptor complex to neuronal viability. In the developing retina, Sortilin 1 (Sort1)(-/-) mice showed reduced neuronal apoptosis that was indistinguishable from that observed in p75(NTR)-deficient (Ngfr(-/-)) mice. To our surprise, although sortilin deficiency did not affect developmentally regulated apoptosis of sympathetic neurons, it did prevent their age-dependent degeneration. Furthermore, in an injury protocol, lesioned corticospinal neurons in Sort1(-/-) mice were protected from death. Thus, the sortilin pathway has distinct roles in pro-neurotrophin-induced apoptotic signaling in pathological conditions, but also in specific stages of neuronal development and aging.
Collapse
Affiliation(s)
- Pernille Jansen
- MIND Center, Department of Medical Biochemistry, Ole Worms Allé 1170, Aarhus University, DK-8000 Aarhus C, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Gashegu J, Ladha R, Vanmuylder N, Philippson C, Bremer F, Rooze M, Louryan S. HSP110, caspase-3 and -9 expression in physiological apoptosis and apoptosis induced by in vivo embryonic exposition to all-trans retinoic acid or irradiation during early mouse eye development. J Anat 2007; 210:532-41. [PMID: 17451530 PMCID: PMC2375737 DOI: 10.1111/j.1469-7580.2007.00719.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Apoptosis is an essential physiological process in embryonic development. In the developing eye of vertebrates, three periods of developmental apoptosis can be distinguished: early, intermediate and later. Within the apoptosis pathway, caspases play a crucial role. It has also been shown that HSP110 may have a potential role in apoptosis. The aim of this research was to study the expression of HSP110, caspase-3 and -9 in physiological, retinoic- or irradiation-induced apoptosis during early eye development. Seven pregnant C57Bl/6J mice received 80 mg kg(-1) of all-trans retinoic acid mixed with sesame oil. Seven pregnant NMRI mice received 2 Gy irradiation at the same gestational day. Control mice of both strains (seven mice of each) were not submitted to any treatment. Embryos were harvested at 3, 6, 12 and 24 h after exposition, fixed, dehydrated and embedded. Coronal sections (5 microm) were made. Slide staining occurred alternatively using anti-caspase-3, anti-caspase-9 and anti-HSP110 immunohistochemistry. HSP110 and caspase-3 expression presented similar topographic and chronological patterns, whereas expression of HSP110 was more precocious in retinoic acid-treated embryos. After retinoic exposure, caspase-3- and HSP110-positive cells were increased in the region of the optic vesicle. By contrast, after irradiation, caspase-3- and HSP110-positive cells were noticeably increased in the optic vesicle, peri-optical mesoderm but less in lens placode. HSP110 was expressed before caspase-3. By contrast, caspase-9 was expressed by a very small number of cells in the optic vesicle either under physiological or under teratogenic conditions. Thus, it seems that activation of caspase-9 is dispensable in early eye developmental apoptosis.
Collapse
Affiliation(s)
- Julien Gashegu
- Department of Anatomy and Embryology, Université Libre de Bruxelles, Faculty of Medicine, Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
44
|
Bhargava S. Role of nerve growth factor and its receptor in the morphogenesis of neural tube in early chick embryo. Gen Comp Endocrinol 2007; 153:141-6. [PMID: 17418844 DOI: 10.1016/j.ygcen.2007.02.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2006] [Revised: 02/22/2007] [Accepted: 02/24/2007] [Indexed: 12/11/2022]
Abstract
Expression of p75 nerve growth factor receptor (p75(NTR)) in the early neurogenesis of chick embryo showed that nerve growth factor receptor (NGFR) is localized in presumptive neuroectoblast and endoblast in the chick gastrula but not in the mesoblast. By stages 9, 10, and 11, NGFR positive cells were located distinctly in the region where the neural folds converge, meet and fuse. NGFR expression was also seen in developing notochord and somites, wherein the reaction was localized on the cell surfaces. Strong p75(NTR) reaction was seen on the roof of the neural tube where it detaches from the head ectoderm by stage 12. The study revealed that p75(NTR) is co-expressed with NGF in the same developmental stage(s) and in areas, where cell death occurs during neuronal development. Further, when the endogenous levels of NGF signaling were blocked by anti-NGF antibody, abnormalities were observed at the anterior end of the neural tube formation. As a result, embryos showed open neural tubes and a few were bent on one side of the body axis. In a small proportion of embryos, diffused somites were observed. The findings supports and confirms our previous study that NGF signaling plays a significant role in the shaping of neural tube in chick embryos through p75(NTR)-NGF receptor.
Collapse
Affiliation(s)
- Shobha Bhargava
- Molecular Embryology Laboratory, Department of Zoology, University of Pune, Pune 411 007, India.
| |
Collapse
|
45
|
Matveeva NY, Kalinichenko SG, Pushchin II, Motavkin PA. The role of nitric oxide in the apoptosis of neurons in the retina of the human fetal eye. ACTA ACUST UNITED AC 2007; 37:111-8. [PMID: 17187201 DOI: 10.1007/s11055-007-0157-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2005] [Indexed: 12/22/2022]
Abstract
The locations of NADPH-diaphorase (NADPH-d), inducible NO synthase (iNOS), and TUNEL-immunoreactive neurons in the retina of human fetuses collected during the first to third trimesters of pregnancy were studied. High levels of NADPH-d activity were seen in the inner segments of light-sensitive cells, amacrine cells, and ganglion cells. The population of NADPH-d-positive amacrine cells included three types of neuron. Type 1 neurons were large and had sparse dendritic fields occupying the inner nuclear and outer retinal layers. Small type 2 neurons were located in the inner retinal layer. Ectopic amacrine cells, type 3, were located in the outer part of the ganglion layer. A high density of NADPH-d-positive neurons was seen in the central part of the retina, surrounding the central fovea and optic disk area. NADPH-d activity increased progressively during ontogenesis and correlated with the appearance of immunoreactive iNOS in neurons. iNOS labeled a subpopulation of amacrine and ganglion cells, which appeared at 20-21 weeks of development and reached a peak of immunoreactivity by the end of the third trimester. TUNEL-immunopositive neuron nuclei with signs of apoptotic destruction were seen at 30-31 weeks of pregnancy. The greatest apoptotic index was seen in the ganglion and amacrine cell populations. These data identify NO as a factor mediating apoptosis of neurons during the critical period of differentiation of interneuronal connections in the human retina.
Collapse
Affiliation(s)
- N Yu Matveeva
- Faculty of Histology, Vladivostok State Medical University, Vladivostok, Russia
| | | | | | | |
Collapse
|
46
|
Harada T, Harada C, Parada LF. Molecular regulation of visual system development: more than meets the eye. Genes Dev 2007; 21:367-78. [PMID: 17322396 DOI: 10.1101/gad.1504307] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Vertebrate eye development has been an excellent model system to investigate basic concepts of developmental biology ranging from mechanisms of tissue induction to the complex patterning and bidimensional orientation of the highly specialized retina. Recent advances have shed light on the interplay between numerous transcriptional networks and growth factors that are involved in the specific stages of retinogenesis, optic nerve formation, and topographic mapping. In this review, we summarize this recent progress on the molecular mechanisms underlying the development of the eye, visual system, and embryonic tumors that arise in the optic system.
Collapse
Affiliation(s)
- Takayuki Harada
- Department of Developmental Biology, Kent Waldrep Foundation Center for Basic Neuroscience Research on Nerve Growth and Regeneration, University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | | | | |
Collapse
|
47
|
Krieglstein K. Cell death in the nervous system. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 557:1-10. [PMID: 16955701 DOI: 10.1007/0-387-30128-3_1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
48
|
Doherty GH. Developmental switch in the effects of TNFα on ventral midbrain dopaminergic neurons. Neurosci Res 2007; 57:296-305. [PMID: 17150272 DOI: 10.1016/j.neures.2006.10.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2006] [Revised: 10/27/2006] [Accepted: 10/30/2006] [Indexed: 12/22/2022]
Abstract
The cytokine tumour necrosis factor-alpha (TNFalpha) has been implicated in the pathogenesis of neurodegenerative conditions as well as in the establishment of neural networks during development. This study investigated the in vitro effects of TNFalpha on embryonic dopaminergic neurons of the ventral mesencephalon. TNFalpha treatment enhanced the number of dopaminergic neurons in cultures derived from E12.5 mice embryos in a dose-dependent manner. In order to achieve this effect TNFalpha signalled via NF-kappaB. This enhancement in cell number was found to be due to TNFalpha promoting the differentiation of dopaminergic neurons rather than to an increase in cell survival. In contrast, TNFalpha-treated cultures derived from E14 or E16 mice demonstrated a decrease in dopaminergic neurons, and this loss was negated by pharmacological inhibition of caspases. The data presented demonstrate that during embryonic development, dopaminergic ventral mesencephalic neurons switch their in vitro response to TNFalpha from neurotrophic to neurotoxic. This is the first report of a population of neurons exhibiting this switch in TNFalpha responsiveness during neurodevelopment.
Collapse
Affiliation(s)
- G H Doherty
- School of Biology, University of St Andrews, Bute Medical Buildings, St Mary's Quad, St Andrews, Fife, Scotland, UK.
| |
Collapse
|
49
|
Elliott J, Cayouette M, Gravel C. The CNTF/LIF signaling pathway regulates developmental programmed cell death and differentiation of rod precursor cells in the mouse retina in vivo. Dev Biol 2006; 300:583-98. [PMID: 17054938 DOI: 10.1016/j.ydbio.2006.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Revised: 08/31/2006] [Accepted: 09/05/2006] [Indexed: 11/19/2022]
Abstract
Natural cell death is critical for normal development of the nervous system, but the extracellular regulators of developmental cell death remain poorly characterized. Here, we studied the role of the CNTF/LIF signaling pathway during mouse retinal development in vivo. We show that exposure to CNTF during neonatal retinal development in vivo retards rhodopsin expression and results in an important and specific deficit in photoreceptor cells. Detailed analysis revealed that exposure to CNTF during retinal development causes a sharp increase in cell death of postmitotic rod precursor cells. Importantly, we show that blocking the CNTF/LIF signaling pathway during mouse retinal development in vivo results in a significant reduction of naturally occurring cell death. Using retroviral lineage analysis, we demonstrate that exposure to CNTF causes a specific reduction of clones containing only rods without affecting other clone types, whereas blocking the CNTF/LIF receptor complex causes a specific increase of clones containing only rods. In addition, we show that stimulation of the CNTF/LIF pathway positively regulates the expression of the neuronal and endothelial nitric oxide synthase (NOS) genes, and blocking nitric oxide production by pre-treatment with a NOS inhibitor abolishes CNTF-induced cell death. Taken together, these results indicate that the CNTF/LIF signaling pathway acts via regulation of nitric oxide production to modulate developmental programmed cell death of postmitotic rod precursor cells.
Collapse
Affiliation(s)
- Jimmy Elliott
- Institut de Recherches Cliniques de Montréal (IRCM), 110, avenue des Pins Ouest Montréal, Québec, Canada H2W 1R7
| | | | | |
Collapse
|
50
|
Abstract
Neurotrophins are a small family of dimeric secretory proteins in vertebrate neurons with a broad spectrum of functions. They are generated as pro-proteins with a functionality that is distinct from the proteolytically processed form. The cellular responses of neurotrophins are mediated by three different types of receptor proteins, the receptor tyrosine kinases of the Trk family, the neurotrophin receptor p75(NTR), which is a member of the tumor necrosis factor receptor (TNFR) superfamily, and sortilin, previously characterized as neurotensin receptor. Recent studies have revealed an intriguing pattern: neurotrophins can elicit opposing signals utilising their variable configuration and different receptor types.
Collapse
Affiliation(s)
- Rüdiger Schweigreiter
- Biocenter Innsbruck, Division of Neurobiochemistry, Medical University Innsbruck, Fritz-Pregl-Strasse 3, A-6020 Innsbruck Innsbruck, Austria.
| |
Collapse
|