1
|
Landucci E, Mango D, Carloni S, Mazzantini C, Pellegrini-Giampietro DE, Saidi A, Balduini W, Schiavi E, Tigli L, Pioselli B, Imbimbo BP, Facchinetti F. Beneficial effects of CHF6467, a modified human nerve growth factor, in experimental neonatal hypoxic-ischaemic encephalopathy. Br J Pharmacol 2025; 182:510-529. [PMID: 39379341 DOI: 10.1111/bph.17353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/03/2024] [Accepted: 07/13/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND AND PURPOSE Therapeutic hypothermia (TH) has become the standard care to reduce morbidity and mortality in neonates affected by moderate-to-severe hypoxic-ischaemic encephalopathy (HIE). Despite the use of TH for HIE, the incidence of mortality and disabilities remains high. EXPERIMENTAL APPROACH Nerve growth factor (NGF) is a potent neurotrophin, but clinical use is limited by its pain eliciting effects. CHF6467 is a recombinant modified form of human NGF devoid of algogenic activity (painless NGF). KEY RESULTS In rodent hippocampal slices exposed to oxygen and glucose deprivation, CHF6467 protected neurons from death and reverted neurotransmission impairment when combined with hypothermia. In a model of rat neonatal HIE, intranasal CHF6467 (20 μg kg-1) significantly reduced brain infarct volume versus vehicle when delivered 10 min or 3 h after the insult. CHF6467 (20 and 40 μg kg-1, i.n.), significantly decreased brain infarct volume to a similar extent to TH and when combined, showed a synergistic neuroprotective effect. CHF6467 (20 μg kg-1, i.n.) per se and in combination with hypothermia reversed locomotor coordination impairment (Rotarod test) and memory deficits (Y-maze and novel object recognition test) in the neonatal HIE rat model. Intranasal administration of CHF6467 resulted in meaningful concentrations in the brain, blunted HIE-induced mRNA elevation of brain neuroinflammatory markers and, when combined to TH, significantly counteracted the increase in plasma levels of neurofilament light chain, a peripheral marker of neuroaxonal damage. CONCLUSION AND IMPLICATIONS CHF6467 administered intranasally is a promising therapy, in combination with TH, for the treatment of HIE.
Collapse
Affiliation(s)
- Elisa Landucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Dalila Mango
- Laboratory Pharmacology of Synaptic Plasticity, European Brain Research Institute, Rome, Italy
- School of Pharmacy, University of Rome "Tor Vergata", Rome, Italy
| | - Silvia Carloni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Costanza Mazzantini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | | | - Amira Saidi
- Laboratory Pharmacology of Synaptic Plasticity, European Brain Research Institute, Rome, Italy
| | - Walter Balduini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Elisa Schiavi
- Corporate Preclinical R&D, Chiesi Farmaceutici, Parma, Italy
| | - Laura Tigli
- Corporate Preclinical R&D, Chiesi Farmaceutici, Parma, Italy
| | | | - Bruno P Imbimbo
- Corporate Preclinical R&D, Chiesi Farmaceutici, Parma, Italy
| | | |
Collapse
|
2
|
Latini L, De Araujo DSM, Amato R, Canovai A, Buccarello L, De Logu F, Novelli E, Vlasiuk A, Malerba F, Arisi I, Florio R, Asari H, Capsoni S, Strettoi E, Villetti G, Imbimbo BP, Dal Monte M, Nassini R, Geppetti P, Marinelli S, Cattaneo A. A p75 neurotrophin receptor-sparing nerve growth factor protects retinal ganglion cells from neurodegeneration by targeting microglia. Br J Pharmacol 2024; 181:4890-4919. [PMID: 39252503 DOI: 10.1111/bph.17316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/22/2024] [Accepted: 06/10/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND AND PURPOSE Retinal ganglion cells (RGCs) are the output stage of retinal information processing, via their axons forming the optic nerve (ON). ON damage leads to axonal degeneration and death of RGCs, and results in vision impairment. Nerve growth factor (NGF) signalling is crucial for RGC operations and visual functions. Here, we investigate a new neuroprotective mechanism of a novel therapeutic candidate, a p75-less, TrkA-biased NGF agonist (hNGFp) in rat RGC degeneration, in comparison with wild type human NGF (hNGFwt). EXPERIMENTAL APPROACH Both neonate and adult rats, whether subjected or not to ON lesion, were treated with intravitreal injections or eye drops containing either hNGFp or hNGFwt. Different doses of the drugs were administered at days 1, 4 or 7 after injury for a maximum of 10 days, when immunofluorescence, electrophysiology, cellular morphology, cytokine array and behaviour studies were carried out. Pharmacokinetic evaluation was performed on rabbits treated with hNGFp ocular drops. RESULTS hNGFp exerted a potent RGC neuroprotection by acting on microglia cells, and outperformed hNGFwt in rescuing RGC degeneration and reducing inflammatory molecules. Delayed use of hNGFp after ON lesion resulted in better outcomes compared with treatment with hNGFwt. Moreover, hNGFp-based ocular drops were less algogenic than hNGFwt. Pharmacokinetic measurements revealed that biologically relevant quantities of hNGFp were found in the rabbit retina. CONCLUSIONS AND IMPLICATIONS Our data point to microglia as a new cell target through which NGF-induced TrkA signalling exerts neuroprotection of the RGC, emphasizing hNGFp as a powerful treatment to tackle retinal degeneration.
Collapse
Affiliation(s)
- Laura Latini
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | | | - Rosario Amato
- Department of Biology, University of Pisa, Pisa, Italy
| | | | - Lucia Buccarello
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Elena Novelli
- Institute of Neuroscience, Italian National Research Council-CNR, Pisa, Italy
| | - Anastasiia Vlasiuk
- Faculty of Biosciences, Collaboration for Joint PhD Degree Between EMBL and Heidelberg University, Heidelberg, Germany
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Rome, Italy
| | - Francesca Malerba
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Ivan Arisi
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Rita Florio
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Hiroki Asari
- Faculty of Biosciences, Collaboration for Joint PhD Degree Between EMBL and Heidelberg University, Heidelberg, Germany
| | - Simona Capsoni
- BIO@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
- Section of Human Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Enrica Strettoi
- Institute of Neuroscience, Italian National Research Council-CNR, Pisa, Italy
| | - Gino Villetti
- Department of Research & Development, Chiesi Farmaceutici, Parma, Italy
| | | | | | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Silvia Marinelli
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Antonino Cattaneo
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
- BIO@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
| |
Collapse
|
3
|
Morado-Urbina CE, Kato J, Sandor K, Vazquez-Mora JA, Ängeby Möller K, Simon N, Salcido J, Martinez-Martinez A, Munoz-Islas E, Jimenez-Andrade JM, Svensson CI. Sex-dependent effects of the targeted nerve growth factor mutation (R100E) on pain behavior, joint inflammation, and bone erosion in mice. Pain 2024; 165:2814-2828. [PMID: 39324959 PMCID: PMC11562760 DOI: 10.1097/j.pain.0000000000003343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 05/10/2024] [Accepted: 06/07/2024] [Indexed: 09/27/2024]
Abstract
ABSTRACT Nerve growth factor (NGF)-R100E is a mutated form of human recombinant NGF that reduces the binding of NGF to its p75NTR receptor while retaining its affinity toward the TrkA receptor. Here, we used human wild type NGF and NGF-R100E knock-in mice to investigate the effects of this NGF mutation on inflammation-induced pain-related behaviors and bone loss. The hNGF-R100E mutation did not alter the nerve fiber density in the sciatic nerve, ankle joint synovium, and skin of naïve mice. Withdrawal responses to mechanical, thermal, and cold stimuli before and after joint inflammation induced by intra-articular injection of complete Freund adjuvant (CFA) were similar between human recombinant nerve growth factor-wild type and hNGF-R100E male and female mice while weight bearing and gait analysis revealed significant differences. Intriguingly, hNGF-R100E male and female mice showed only mild changes, indicating lower degrees of deep joint-related pain compared to their wild type counterparts. Furthermore, micro-CT analysis demonstrated that hNGF-R100E female mice, but not males, were protected from CFA-induced bone loss, and mRNA analysis showed a different gene regulation indicating a sex-dependent relationship between NGF, inflammation, and bone loss. In conclusion, our study reveals that the hNGF-R100E mutation renders mice insensitive to inflammation-induced impact on joint loading and gait while preserving the development of the peripheral nociceptive neurons and sensitivity to punctate stimulation of the skin. Notably, the mutation uncovers a sex-dependent relationship between NGF and inflammation-induced bone loss. These findings offer valuable insights into NGF as a target for pain management and the interplay between NGF and bone architecture.
Collapse
Affiliation(s)
- Carlos E. Morado-Urbina
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Jungo Kato
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
- Department of Anesthesiology, Keio University School of Medicine, Tokyo, Japan
| | - Katalin Sandor
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Juan Antonio Vazquez-Mora
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Kristina Ängeby Möller
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Nils Simon
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Jaira Salcido
- Unidad Académica Multidisciplinaria Reynosa Aztlán, Universidad Autónoma de Tamaulipas, Reynosa, México
| | - Arisai Martinez-Martinez
- Unidad Académica Multidisciplinaria Reynosa Aztlán, Universidad Autónoma de Tamaulipas, Reynosa, México
| | - Enriqueta Munoz-Islas
- Unidad Académica Multidisciplinaria Reynosa Aztlán, Universidad Autónoma de Tamaulipas, Reynosa, México
| | | | - Camilla I. Svensson
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
4
|
Silvestri F, Montuoro R, Catalani E, Tilesi F, Willems D, Romano N, Ricciardi S, Cervia D, Ceci M. eIF3d specialized translation requires a RACK1-driven eIF3d binding to 43S PIC in proliferating SH-SY5Y neuroblastoma cells. Cell Signal 2024; 125:111494. [PMID: 39477045 DOI: 10.1016/j.cellsig.2024.111494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/19/2024] [Accepted: 10/27/2024] [Indexed: 11/05/2024]
Abstract
Translation initiation of most mammalian mRNAs is mediated by a 5' cap structure that binds eukaryotic initiation factor 4E (eIF4E). Notably, most mRNAs are still capped when eIF4E is inhibited, suggesting alternative mechanisms likely mediate cap-dependent mRNA translation without functional eIF4F. Here we found that, when eIF4E is inhibited, the ribosomal scaffold RACK1 recruits eIF3d on the 43S pre-initiation complex. Moreover, we found that it is just PKCBII in its active form that promotes the binding of RACK1 to eIF3d. These studies disclose a previously unknown role of ribosomal RACK1 for eIF3d specialized translation.
Collapse
Affiliation(s)
- Federica Silvestri
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy.
| | - Raffaele Montuoro
- Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy.
| | - Francesca Tilesi
- Department of Ecological and Biological Science (DEB), Università degli Studi Della Tuscia, Viterbo, Italy.
| | - Daniela Willems
- Department of Ecological and Biological Science (DEB), Università degli Studi Della Tuscia, Viterbo, Italy.
| | - Nicla Romano
- Department of Ecological and Biological Science (DEB), Università degli Studi Della Tuscia, Viterbo, Italy.
| | - Sara Ricciardi
- National Institute of Molecular Genetics, INGM "Romeo ed Enrica Invernizzi", 20122 Milan, Italy; Department of Biological Sciences, DBS, University of Milan, 20133 Milan, Italy.
| | - Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy.
| | - Marcello Ceci
- Department of Ecological and Biological Science (DEB), Università degli Studi Della Tuscia, Viterbo, Italy.
| |
Collapse
|
5
|
Napoli D, Orsini N, Salamone G, Calvello MA, Capsoni S, Cattaneo A, Strettoi E. Human NGF "Painless" Ocular Delivery for Retinitis Pigmentosa: An In Vivo Study. eNeuro 2024; 11:ENEURO.0096-24.2024. [PMID: 39293937 PMCID: PMC11412101 DOI: 10.1523/eneuro.0096-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 09/20/2024] Open
Abstract
Retinitis pigmentosa (RP) is a family of genetically heterogeneous diseases still without a cure. Despite the causative genetic mutation typically not expressed in cone photoreceptors, these cells inevitably degenerate following the primary death of rods, causing blindness. The reasons for the "bystander" degeneration of cones are presently unknown but decrement of survival factors, oxidative stress, and inflammation all play a role. Targeting these generalized biological processes represents a strategy to develop mutation-agnostic therapies for saving vision in large populations of RP individuals. A classical method to support neuronal survival is by employing neurotrophic factors, such as NGF. This study uses painless human NGF (hNGFp), a TrkA receptor-biased variant of the native molecule with lower affinity for nociceptors and limited activity as a pain inducer; the molecule has identical neurotrophic power of the native form but a reduced affinity for the p75NTR receptors, known to trigger apoptosis. hNGFp has a recognized activity on brain microglial cells, which are induced to a phenotype switch from a highly activated to a more homeostatic configuration. hNGFp was administered to RP-like mice in vivo with the aim of decreasing retinal inflammation and also providing retinal neuroprotection. However, the ability of this treatment to counteract the bystander degeneration of cones remained limited.
Collapse
Affiliation(s)
- Debora Napoli
- CNR Neuroscience Institute, Pisa 56124, Italy
- Regional Doctorate School in Neuroscience, University of Florence, Italy
| | - Noemi Orsini
- CNR Neuroscience Institute, Pisa 56124, Italy
- Regional Doctorate School in Neuroscience, University of Florence, Italy
| | | | | | - Simona Capsoni
- Section of Human Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy
| | - Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
- Rita Levi-Montalcini European Brain Research Institute (EBRI), Roma 00161, Italy
| | | |
Collapse
|
6
|
Liu A, Mohr MA, Hope JM, Wang J, Chen X, Cui B. Light-Inducible Activation of TrkA for Probing Chronic Pain in Mice. ACS Chem Biol 2024; 19:1626-1637. [PMID: 39026469 PMCID: PMC11756861 DOI: 10.1021/acschembio.4c00300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Chronic pain is a prevalent problem that plagues modern society, and better understanding its mechanisms is critical for developing effective therapeutics. Nerve growth factor (NGF) and its primary receptor, Tropomyosin receptor kinase A (TrkA), are known to be potent mediators of chronic pain, but there is a lack of established methods for precisely perturbing the NGF/TrkA signaling pathway in the study of pain and nociception. Optobiological tools that leverage light-induced protein-protein interactions allow for precise spatial and temporal control of receptor signaling. Previously, our lab reported a blue light-activated version of TrkA generated using light-induced dimerization of the intracellular TrkA domain, opto-iTrkA. In this work, we show that opto-iTrkA activation is able to activate endogenous ERK and Akt signaling pathways and causes the retrograde transduction of phospho-ERK signals in dorsal root ganglion (DRG) neurons. Opto-iTrkA activation also sensitizes the transient receptor potential vanilloid 1 (TRPV1) channel in cellular models, further corroborating the physiological relevance of the optobiological stimulus. Finally, we show that opto-iTrkA enables light-inducible potentiation of mechanical sensitization in mice. Light illumination enables nontraumatic and reversible (<2 days) sensitization of mechanical pain in mice transduced with opto-iTrkA, which provides a platform for dissecting TrkA pathways for nociception in vitro and in vivo.
Collapse
Affiliation(s)
- Aofei Liu
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Manuel A Mohr
- Department of Biology, Stanford University, Stanford, California 94305, United States
| | - Jen M Hope
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Jennifer Wang
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Xiaoke Chen
- Department of Biology, Stanford University, Stanford, California 94305, United States
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
7
|
Tiberi A, Borgonovo G, Testa G, Pacifico P, Jacob A, Di Caprio M, Totaro V, Calvello M, Cattaneo A, Capsoni S. Reversal of neurological deficits by painless nerve growth factor in a mouse model of Rett syndrome. Brain 2024; 147:122-134. [PMID: 37633263 PMCID: PMC10766238 DOI: 10.1093/brain/awad282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 07/12/2023] [Accepted: 07/31/2023] [Indexed: 08/28/2023] Open
Abstract
Rett syndrome is a rare genetic neurodevelopmental disease, affecting 1 in over 10 000 females born worldwide, caused by de novo mutations in the X-chromosome-located methyl-CpG-binding protein 2 (MeCP2) gene. Despite the great effort put forth by the scientific community, a therapy for this devastating disease is still needed. Here, we tested the therapeutic effects of a painless mutein of the nerve growth factor (NGF), called human NGF painless (hNGFp), via a non-invasive intranasal delivery in female MeCP2+/- mice. Of note, previous work had demonstrated a broad biodistribution of hNGFp in the mouse brain by the nasal delivery route. We report that (i) the long-term lifelong treatment of MeCP2+/- mice with hNGFp, starting at 2 months of age, increased the chance of survival while also greatly improving behavioural parameters. Furthermore, when we assessed the phenotypic changes brought forth by (ii) a short-term 1-month-long hNGFp-treatment, starting at 3 months of age (right after the initial presentation of symptoms), we observed the rescue of a well known neuronal target population of NGF, cholinergic neurons in the medial septum. Moreover, we reveal a deficit in microglial morphology in MeCP2+/- mice, completely reversed in treated animals. This effect on microglia is in line with reports showing microglia to be a TrkA-dependent non-neuronal target cell population of NGF in the brain. To understand the immunomodulatory activity of hNGFp, we analysed the cytokine profile after hNGFp treatment in MeCP2+/- mice, to discover that the treatment recovered the altered expression of key neuroimmune-communication molecules, such as fractalkine. The overall conclusion is that hNGFp delivered intranasally can ameliorate symptoms in the MeCP2+/- model of Rett syndrome, by exerting strong neuroprotection with a dual mechanism of action: directly on target neurons and indirectly via microglia.
Collapse
Affiliation(s)
- Alexia Tiberi
- Institute of Neuroscience, CNR, 56124 Pisa, Italy
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Giulia Borgonovo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Giovanna Testa
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Paola Pacifico
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Ajesh Jacob
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | | | - Valentino Totaro
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | | | - Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
- Rita Levi-Montalcini European Brain Research Institute (EBRI), 00161 Roma, Italy
| | - Simona Capsoni
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
- Section of Human Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
8
|
Tringali G, Pizzoferrato M, Lisi L, Marinelli S, Buccarello L, Falsini B, Cattaneo A, Navarra P. A Vicious NGF-p75 NTR Positive Feedback Loop Exacerbates the Toxic Effects of Oxidative Damage in the Human Retinal Epithelial Cell Line ARPE-19. Int J Mol Sci 2023; 24:16237. [PMID: 38003427 PMCID: PMC10671591 DOI: 10.3390/ijms242216237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
In spite of its variety of biological activities, the clinical exploitation of human NGF (hNGF) is currently limited to ocular pathologies. It is therefore interesting to test the effects of hNGF in preclinical models that may predict their efficacy and safety in the clinical setting of ocular disorders and compare the effects of hNGF with those of its analogs. We used a human retinal pigment cell line, ARPE-19 cells, to investigate the effects of hNGF and its analogs, mouse NGF (mNGF) and painless NGF (pNGF), on cell viability under basal conditions and after exposure to oxidative stimuli, i.e., hydrogen peroxide (H2O2) and ultraviolet (UV)-A rays. The effects of hNGF and pNGF were also tested on the gene expression and protein synthesis of the two NGF receptor subtypes, p75 neurotrophic receptors (p75NTR) and tyrosine kinase A (TrkA) receptors. We drew the following conclusions: (i) the exposure of ARPE-19 cells to H2O2 or UV-A causes a dose-dependent decrease in the number of viable cells; (ii) under baseline conditions, hNGF, but not pNGF, causes a concentration-dependent decrease in cell viability in the range of doses 1-100 ng/mL; (iii) hNGF, but not pNGF, significantly potentiates the toxic effects of H2O2 or of UV-A on ARPE-19 cells in the range of doses 1-100 ng/mL, while mNGF at the same doses presents an intermediate behavior; (iv) 100 ng/mL of hNGF triggers an increase in p75NTR expression in H2O2-treated ARPE-19 cells, while pNGF at the same dose does not; (v) pNGF, but not hNGF (both given at 100 ng/mL), increases the total cell fluorescence intensity for TrkA receptors in H2O2-treated ARPE-19 cells. The present findings suggest a vicious positive feedback loop through which NGF-mediated upregulation of p75NTR contributes to worsening the toxic effects of oxidative damage in the human retinal epithelial cell line ARPE-19. Looking at the possible clinical relevance of these findings, one can postulate that pNGF might show a better benefit/risk ratio than hNGF in the treatment of ocular disorders.
Collapse
Affiliation(s)
- Giuseppe Tringali
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy (M.P.)
| | - Michela Pizzoferrato
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy (M.P.)
| | - Lucia Lisi
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy (M.P.)
| | - Silvia Marinelli
- European Brain Research Institute-Fondazione Rita Levi Montalcini, 00161 Rome, Italy (L.B.)
| | - Lucia Buccarello
- European Brain Research Institute-Fondazione Rita Levi Montalcini, 00161 Rome, Italy (L.B.)
| | - Benedetto Falsini
- UOC Ophtalmology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Department of Ophthalmology, Bambino Gesù IRCCS Children’s Hospital, 00133 Rome, Italy
| | - Antonino Cattaneo
- European Brain Research Institute-Fondazione Rita Levi Montalcini, 00161 Rome, Italy (L.B.)
- Bio@SNS Laboratory, Scuola Normale Superiore, 56124 Pisa, Italy
| | - Pierluigi Navarra
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy (M.P.)
| |
Collapse
|
9
|
Covaceuszach S, Lamba D. The NGF R100W Mutation, Associated with Hereditary Sensory Autonomic Neuropathy Type V, Specifically Affects the Binding Energetic Landscapes of NGF and of Its Precursor proNGF and p75NTR. BIOLOGY 2023; 12:biology12030364. [PMID: 36979056 PMCID: PMC10045213 DOI: 10.3390/biology12030364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023]
Abstract
Nerve Growth Factor (NGF), the prototype of the neurotrophin family, stimulates morphological differentiation and regulates neuronal gene expression by binding to TrkA and p75NTR receptors. It plays a critical role in maintaining the function and phenotype of peripheral sensory and sympathetic neurons and in mediating pain transmission and perception during adulthood. A point mutation in the NGFB gene (leading to the amino acid substitution R100W) is responsible for Hereditary Sensory and Autonomic Neuropathy type V (HSAN V), leading to a congenital pain insensitivity with no clear cognitive impairments, but with alterations in the NGF/proNGF balance. The available crystal structures of the p75NTR/NGF and 2p75NTR/proNGF complexes offer a starting point for Molecular Dynamics (MD) simulations in order to capture the impact of the R100W mutation on their binding energetic landscapes and to unveil the molecular determinants that trigger their different physiological and pathological outcomes. The present in silico studies highlight that the stability and the binding energetic fingerprints in the 2p75NTR/proNGF complex is not affected by R100W mutation, which on the contrary, deeply affects the energetic landscape, and thus the stability in the p75NTR/NGF complex. Overall, these findings present insights into the structural basis of the molecular mechanisms beyond the clinical manifestations of HSAN V patients.
Collapse
Affiliation(s)
- Sonia Covaceuszach
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, 34149 Trieste, Italy
- Correspondence: (S.C.); (D.L.)
| | - Doriano Lamba
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, 34149 Trieste, Italy
- Consorzio Interuniversitario “Istituto Nazionale Biostrutture e Biosistemi”, 00136 Roma, Italy
- Correspondence: (S.C.); (D.L.)
| |
Collapse
|
10
|
Yu X, Yang Z, Zhang Y, Xia J, Zhang J, Han Q, Yu H, Wu C, Xu Y, Xu W, Yang W. Lipid Nanoparticle Delivery of Chemically Modified NGF R100W mRNA Alleviates Peripheral Neuropathy. Adv Healthc Mater 2023; 12:e2202127. [PMID: 36325948 DOI: 10.1002/adhm.202202127] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/26/2022] [Indexed: 11/06/2022]
Abstract
Messenger RNA (mRNA) carries genetic instructions to the cell machinery for the transient production of antigens or therapeutic proteins and shows enormous potential in vaccine development, cancer immunotherapy, protein replacement therapy, and genome engineering. Here, the synthesis of chemically modified nerve growth factor mutant (NGFR100W ) mRNA through in vitro transcription is described. After the replacement of the original signal peptide sequence with the Ig Kappa leader sequence, codon-optimized NGFR100W mRNA yielded high secretion of mature NGFR100W , which promotes axon growth in PC12 cells. Using lipid nanoparticle (LNP)-delivery of N1-methylpseudouridine-modified mRNA in mice, NGFR100W -mRNA-LNPs result in the successful expression of NGFR100W protein, which significantly reduces nociceptive activity compared to that of NGFWT . This indicates that NGFR100W derived from exogenous mRNA elicited "painless" neuroprotective activity. Additionally, the therapeutic value of NGFR100W mRNA is established in a paclitaxel-induced peripheral neuropathy model by demonstrating the rapid recovery of intraepidermal nerve fibers. The results show that in vitro-transcribed mRNA has significant flexibility in sequence design and fast in vivo functional validation of target proteins. Furthermore, the results highlight the therapeutic potential of mRNA as a supplement to beneficial proteins for preventing or reversing some chronic medical conditions, such as peripheral neuropathy.
Collapse
Affiliation(s)
- Xiang Yu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Zheng Yang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Yu Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Jia Xia
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China
| | - Jiahui Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Qi Han
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Hang Yu
- Shanghai RNACure Biopharma Co., Ltd., Shanghai, 200438, P. R. China
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92037, USA
| | - Yingjie Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China.,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Wei Xu
- Department of Neurology, Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China.,Department of Neurology, Ruijin Hospital, Zhoushan Branch, Shanghai Jiaotong University School of Medicine, Shanghai, 316012, P. R. China
| | - Wen Yang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai, 200025, P. R. China
| |
Collapse
|
11
|
Testa G, Mainardi M, Vannini E, Pancrazi L, Cattaneo A, Costa M. Disentangling the signaling complexity of nerve growth factor receptors by
CRISPR
/Cas9. FASEB J 2022; 36:e22498. [PMID: 37036720 DOI: 10.1096/fj.202101760rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 07/25/2022] [Accepted: 08/02/2022] [Indexed: 11/11/2022]
Abstract
The binding of nerve growth factor (NGF) to the tropomyosin-related kinase A (TrkA) and p75NTR receptors activates a large variety of pathways regulating critical processes as diverse as proliferation, differentiation, membrane potential, synaptic plasticity, and pain. To ascertain the details of TrkA-p75NTR interaction and cooperation, a plethora of experiments, mostly based on receptor overexpression or downregulation, have been performed. Among the heterogeneous cellular systems used for studying NGF signaling, the PC12 pheochromocytoma-derived cell line is a widely used model. By means of CRISPR/Cas9 genome editing, we created PC12 cells lacking TrkA, p75NTR , or both. We found that TrkA-null cells become unresponsive to NGF. Conversely, the absence of p75NTR enhances the phosphorylation of TrkA and its effectors. Using a patch-clamp, we demonstrated that the individual activation of TrkA and p75NTR by NGF results in antagonizing effects on the membrane potential. These newly developed PC12 cell lines can be used to investigate the specific roles of TrkA and p75NTR in a genetically defined cellular model, thus providing a useful platform for future studies and further gene editing.
Collapse
Affiliation(s)
- Giovanna Testa
- Laboratory of Biology “Bio@SNS” Scuola Normale Superiore Pisa Italy
| | - Marco Mainardi
- Laboratory of Biology “Bio@SNS” Scuola Normale Superiore Pisa Italy
- Neuroscience Institute National Research Council (CNR) Pisa Italy
| | - Eleonora Vannini
- Neuroscience Institute National Research Council (CNR) Pisa Italy
| | - Laura Pancrazi
- Neuroscience Institute National Research Council (CNR) Pisa Italy
| | - Antonino Cattaneo
- Laboratory of Biology “Bio@SNS” Scuola Normale Superiore Pisa Italy
- European Brain Research Institute “Rita Levi Montalcini” (EBRI) Rome Italy
| | - Mario Costa
- Laboratory of Biology “Bio@SNS” Scuola Normale Superiore Pisa Italy
- Neuroscience Institute National Research Council (CNR) Pisa Italy
- Centro Pisano ricerca e implementazione clinica Flash Radiotherapy “CPFR@CISUP”, “S. Chiara” Hospital Pisa Italy
| |
Collapse
|
12
|
Lisi L, Marinelli S, Ciotti GMP, Pizzoferrato M, Palmerio F, Chiavari M, Cattaneo A, Navarra P. The effects of painless nerve growth factor on human microglia polarization. Front Cell Neurosci 2022; 16:969058. [PMID: 36339818 PMCID: PMC9633670 DOI: 10.3389/fncel.2022.969058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/03/2022] [Indexed: 11/26/2023] Open
Abstract
Previous studies in the rat suggest that microglial cells represent a potential druggable target for nerve growth factor (NGF) in the brain. The painless human Nerve Growth Factor (hNGFp) is a recombinant mutated form of human nerve growth factor (hNGF) that shows identical neurotrophic and neuroprotective properties of wild-type NGF but displays at least 10-fold lower algogenic activity. From the pharmacological point of view, hNGFp is a biased tropomyosin receptor kinase A (TrkA) agonist and displays a significantly lower affinity for the p75 neurotrophin receptor (p75NTR). This study aimed to evaluate the expression of TrkA and p75NTR NGF receptors in two different human microglia cell lines, and to investigate the effects of hNGFp and wild-type NGF (NGF) on L-arginine metabolism, taken as a marker of microglia polarization. Both NGF receptors are expressed in human microglia cell lines and are effective in transducing signals triggered by NGF and hNGFp. The latter and, to a lesser extent, NGF inhibit cytokine-stimulated inducible nitric oxide synthase (iNOS) expression and nitric oxide (NO) production in these cells. Conversely NGF but not hNGFp stimulates arginase-mediated urea production.
Collapse
Affiliation(s)
- Lucia Lisi
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Silvia Marinelli
- European Brain Research Institute-Fondazione Rita Levi Montalcini, Rome, Italy
| | - Gabriella Maria Pia Ciotti
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Michela Pizzoferrato
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Federica Palmerio
- European Brain Research Institute-Fondazione Rita Levi Montalcini, Rome, Italy
| | - Marta Chiavari
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Antonino Cattaneo
- European Brain Research Institute-Fondazione Rita Levi Montalcini, Rome, Italy
- Bio@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
| | - Pierluigi Navarra
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| |
Collapse
|
13
|
Lisi L, Ciotti GMP, Chiavari M, Martire M, Navarra P. The effects of CHF6467, a new mutated form of NGF, on cell models of human glioblastoma. A comparison with wild-type NGF. Growth Factors 2022; 40:37-45. [PMID: 35442129 DOI: 10.1080/08977194.2022.2060095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
CHF6467 is a mutated form of human recombinant nerve growth factor (NGF). The mutation selectively disrupts the binding of NGF to its p75NTR receptor while maintaining the affinity toward TrkA receptor. Because of such different profile of receptor interaction, CHF6467 maintains unaltered the neurotrophic and neuroprotective properties of wild-type NGF but shows reduced algogenic activity.In this study, we investigated the effects of CHF6467 on mortality, proliferation, cell-damage and migration in three human glioblastoma cell lines (U87MG, T98G, LN18), and in the rat astrocytoma C6 cells. Both CHF6467 and wild-type NGF, given in the range 1-50 ng/ml, did not modify cell proliferation, metabolism and migration, as well as the number of live/dead cells.The present in vitro data are predictive of a lack of tumorigenic activity by both wild-type NGF and CHF6467 on these cell types in vivo, and warrant for CHF6467 further clinical development.
Collapse
Affiliation(s)
- Lucia Lisi
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Gabriella Maria Pia Ciotti
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Marta Chiavari
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Maria Martire
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Pierluigi Navarra
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| |
Collapse
|
14
|
Neurotrophin Signaling Impairment by Viral Infections in the Central Nervous System. Int J Mol Sci 2022; 23:ijms23105817. [PMID: 35628626 PMCID: PMC9146244 DOI: 10.3390/ijms23105817] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
Neurotrophins, such as nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin 3 (NT-3), NT-4, and NT-5, are proteins involved in several important functions of the central nervous system. The activation of the signaling pathways of these neurotrophins, or even by their immature form, pro-neurotrophins, starts with their recognition by cellular receptors, such as tropomyosin receptor kinase (Trk) and 75 kD NT receptors (p75NTR). The Trk receptor is considered to have a high affinity for attachment to specific neurotrophins, while the p75NTR receptor has less affinity for attachment with neurotrophins. The correct functioning of these signaling pathways contributes to proper brain development, neuronal survival, and synaptic plasticity. Unbalanced levels of neurotrophins and pro-neurotrophins have been associated with neurological disorders, illustrating the importance of these molecules in the central nervous system. Furthermore, reports have indicated that viruses can alter the normal levels of neurotrophins by interfering with their signaling pathways. This work discusses the importance of neurotrophins in the central nervous system, their signaling pathways, and how viruses can affect them.
Collapse
|
15
|
OUP accepted manuscript. Brain 2022; 145:2250-2275. [DOI: 10.1093/brain/awac096] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 11/13/2022] Open
|
16
|
Manni L, Conti G, Chiaretti A, Soligo M. Intranasal Delivery of Nerve Growth Factor in Neurodegenerative Diseases and Neurotrauma. Front Pharmacol 2021; 12:754502. [PMID: 34867367 PMCID: PMC8635100 DOI: 10.3389/fphar.2021.754502] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/01/2021] [Indexed: 01/01/2023] Open
Abstract
Since the 1980s, the development of a pharmacology based on nerve growth factor (NGF) has been postulated for the therapy of Alzheimer’s disease (AD). This hypothesis was based on the rescuing effect of the neurotrophin on the cholinergic phenotype of the basal forebrain neurons, primarily compromised during the development of AD. Subsequently, the use of NGF was put forward to treat a broader spectrum of neurological conditions affecting the central nervous system, such as Parkinson’s disease, degenerative retinopathies, severe brain traumas and neurodevelopmental dysfunctions. While supported by solid rational assumptions, the progress of a pharmacology founded on these hypotheses has been hampered by the difficulty of conveying NGF towards the brain parenchyma without resorting to invasive and risky delivery methods. At the end of the last century, it was shown that NGF administered intranasally to the olfactory epithelium was able to spread into the brain parenchyma. Notably, after such delivery, pharmacologically relevant concentration of exogenous NGF was found in brain areas located at considerable distances from the injection site along the rostral-caudal axis. These observations paved the way for preclinical characterization and clinical trials on the efficacy of intranasal NGF for the treatment of neurodegenerative diseases and of the consequences of brain trauma. In this review, a summary of the preclinical and clinical studies published to date will be attempted, as well as a discussion about the mechanisms underlying the efficacy and the possible development of the pharmacology based on intranasal conveyance of NGF to the brain.
Collapse
Affiliation(s)
- Luigi Manni
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| | - Giorgio Conti
- Department of Emergency, Intensive Pediatric Therapy and Pediatric Trauma Center, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Chiaretti
- Department of Woman and Child Health, Institute of Pediatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marzia Soligo
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| |
Collapse
|
17
|
Testa G, Cattaneo A, Capsoni S. Understanding pain perception through genetic painlessness diseases: The role of NGF and proNGF. Pharmacol Res 2021; 169:105662. [PMID: 34000361 DOI: 10.1016/j.phrs.2021.105662] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/23/2021] [Accepted: 05/03/2021] [Indexed: 01/25/2023]
Abstract
Nerve growth factor (NGF), by binding to TrkA and p75NTR receptors, regulates the survival and differentiation of sensory neurons during development and mediates pain transmission and perception during adulthood, by acting at different levels of the nervous system. Key to understanding the role of NGF as a pain mediator is the finding that mutations (namely, R121W, V232fs and R221W) in the NGF gene cause painlessness disease Hereditary Sensory and Autonomic Neuropathy type V (HSAN V). Here we shall review the consequences of these NGF mutations, each of which results in specific clinical signs: R221W determines congenital pain insensitivity with no overt cognitive disabilities, whereas V232fs and R121W also result in intellectual disability, thus showing similarities to HSAN IV, which is caused by mutations in TrkA, rather than to HSAN V. Comparing the cellular, biochemical and clinical findings of these mutations could help in better understanding not only the possible mechanisms underlying HSAN V, but also mechanisms of NGF signalling and roles. These mutations alter the balance between NGF and proNGF in favour of an accumulation of the latter, suggesting a possible role of proNGF as a molecule with an analgesic role. Furthermore, the neurotrophic and pronociceptive functions of NGF are split by the R221W mutation, making NGF variants based on this mutation interesting for designing therapeutic applications for many diseases. This review emphasizes the possibility of using the mutations involved in "painlessness" clinical disorders as an innovative approach to identify new proteins and pathways involved in pain transmission and perception. OUTSTANDING QUESTIONS: Why do homozygous HSAN V die postnatally? What is the cause of this early postnatal lethality? Is the development of a mouse or a human feeling less pain affecting higher cognitive and perceptual functions? What is the consequence of the HSAN V mutation on the development of joints and bones? Are the multiple fractures observed in HSAN V patients due exclusively to the carelessness consequent to not feeling pain, or also to an intrinsic frailty of their bones? Are heterodimers of NGFWT and NGFR221W in the heterozygote state formed? And if so, what are the properties of these heterodimeric proteins? How is the processing of proNGFR221W to NGFR221W affected by the mutation?
Collapse
Affiliation(s)
- Giovanna Testa
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
| | - Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy.
| | - Simona Capsoni
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy; Section of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
18
|
Dlamini Z, Mathabe K, Padayachy L, Marima R, Evangelou G, Syrigos KN, Bianchi A, Lolas G, Hull R. Many Voices in a Choir: Tumor-Induced Neurogenesis and Neuronal Driven Alternative Splicing Sound Like Suspects in Tumor Growth and Dissemination. Cancers (Basel) 2021; 13:cancers13092138. [PMID: 33946706 PMCID: PMC8125307 DOI: 10.3390/cancers13092138] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/16/2021] [Accepted: 04/24/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Significant progress has recently been made in understanding the role of the neuronal system in cancer biology, in many solid tumors such as prostate, breast, pancreatic, gastric and brain cancers. Solid tumors and the nervous system appear to influence each other’s development both directly and indirectly. A recurring element in such interactions is constituted by nerve-related substances such as neurotransmitters and neurotrophins, to which the first part of the current review is devoted. The second part of the review focuses on the potential role played by alternative splicing in cancer progression associated with neural signaling. Alternative splicing is the process where pre-mRNA is cut and re-ligated in different ways to give rise to multiple protein isoforms whose expression profile is often cancer specific. Alternative splicing is known to take place in the mRNA of genes that code for proteins involved in neuronal development and the creation of new nerve fibers. The change in alternative splicing patterns that occur as tumors develop and progress may make these splice variants potential targets for the development of drug treatments. They may also serve as diagnostic or prognostic biomarkers. Abstract During development, as tissues expand and grow, they require circulatory, lymphatic, and nervous system expansion for proper function and support. Similarly, as tumors arise and develop, they also require the expansion of these systems to support them. While the contribution of blood and lymphatic systems to the development and progression of cancer is well known and is targeted with anticancer drugs, the contribution of the nervous system is less well studied and understood. Recent studies have shown that the interaction between neurons and a tumor are bilateral and promote metastasis on one hand, and the formation of new nerve structures (neoneurogenesis) on the other. Substances such as neurotransmitters and neurotrophins being the main actors in such interplay, it seems reasonable to expect that alternative splicing and the different populations of protein isoforms can affect tumor-derived neurogenesis. Here, we report the different, documented ways in which neurons contribute to the development and progression of cancer and investigate what is currently known regarding cancer-neuronal interaction in several specific cancer types. Furthermore, we discuss the incidence of alternative splicing that have been identified as playing a role in tumor-induced neoneurogenesis, cancer development and progression. Several examples of changes in alternative splicing that give rise to different isoforms in nerve tissue that support cancer progression, growth and development have also been investigated. Finally, we discuss the potential of our knowledge in alternative splicing to improve tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Zodwa Dlamini
- SAMRC Precision Prevention and Novel Drug Targets for HIV-Associated Cancers (PPNDTHAC) Unit, Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (K.M.); (L.P.); (R.M.); (G.L.); (R.H.)
- Correspondence:
| | - Kgomotso Mathabe
- SAMRC Precision Prevention and Novel Drug Targets for HIV-Associated Cancers (PPNDTHAC) Unit, Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (K.M.); (L.P.); (R.M.); (G.L.); (R.H.)
- Department of Urology, University of Pretoria, Pretoria 0084, South Africa
| | - Llewellyn Padayachy
- SAMRC Precision Prevention and Novel Drug Targets for HIV-Associated Cancers (PPNDTHAC) Unit, Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (K.M.); (L.P.); (R.M.); (G.L.); (R.H.)
- Department of Neurosurgery, University of Pretoria, Pretoria 0084, South Africa
| | - Rahaba Marima
- SAMRC Precision Prevention and Novel Drug Targets for HIV-Associated Cancers (PPNDTHAC) Unit, Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (K.M.); (L.P.); (R.M.); (G.L.); (R.H.)
| | - George Evangelou
- 3rd Department of Medicine, National & Kapodistrian University of Athens, 11527 Athens, Greece; (G.E.); (K.N.S.)
| | - Konstantinos N. Syrigos
- 3rd Department of Medicine, National & Kapodistrian University of Athens, 11527 Athens, Greece; (G.E.); (K.N.S.)
| | | | - Georgios Lolas
- SAMRC Precision Prevention and Novel Drug Targets for HIV-Associated Cancers (PPNDTHAC) Unit, Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (K.M.); (L.P.); (R.M.); (G.L.); (R.H.)
- 3rd Department of Medicine, National & Kapodistrian University of Athens, 11527 Athens, Greece; (G.E.); (K.N.S.)
| | - Rodney Hull
- SAMRC Precision Prevention and Novel Drug Targets for HIV-Associated Cancers (PPNDTHAC) Unit, Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (K.M.); (L.P.); (R.M.); (G.L.); (R.H.)
| |
Collapse
|
19
|
Malerba F, Bruni Ercole B, Florio R, Cattaneo A. A Quantitative Bioassay to Determine the Inhibitory Potency of NGF-TrkA Antagonists. SLAS DISCOVERY 2021; 26:823-830. [PMID: 33874771 DOI: 10.1177/24725552211000672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this article, we demonstrate and validate a new bioassay named the NTAB [NGF-TrkA (nerve growth factor-tropomyosin receptor kinase A) antagonist bioassay] for the determination of the inhibitory potency of NGF-TrkA antagonists, based on the inhibition of NGF-dependent proliferation of the human TF1 erythroleukemic cell line.It is well known that NGF holds great therapeutic potential due to its neurotrophic and neuroprotective properties. NGF is also involved in some pathways, however, principally driven by TrkA that, if not correctly regulated, can lead to unwanted pathological outcomes linked to pain, angiogenesis, and cancer.Indeed, there is an increasing interest, from a therapeutic perspective, in designing new effective molecules (antibodies, antibody fragments, or small molecules) able to inhibit the undesired NGF-TrkA pathway. For these reasons, there is an interest to develop functional cell-based assays for determination of the inhibition potency of compounds inhibiting the NGF-TrkA axis. The NTAB presents significant advantages over other published NGF-TrkA functional bioassays, for these reasons: (1) It is quantitative, (2) it measures a pure TrkA response, (3) it is simpler, (4) it is based on a natural biological response, and (5) it is easily scalable from a lab scale to an automated industrial assay.The NTAB assay was validated with a panel of well-characterized NGF-TrkA inhibitors, yielding characteristic dose-response curves, from which the relative strength of the inhibitors was quantitatively determined and used for comparisons. This new bioassay will be very useful to assist in the validation and prioritization of the best inhibitors among a large number of candidates.
Collapse
Affiliation(s)
- Francesca Malerba
- Fondazione EBRI (European Brain Research Institute) Rita Levi-Montalcini, Rome, Italy
| | - Bruno Bruni Ercole
- Fondazione EBRI (European Brain Research Institute) Rita Levi-Montalcini, Rome, Italy
| | - Rita Florio
- Fondazione EBRI (European Brain Research Institute) Rita Levi-Montalcini, Rome, Italy
| | - Antonino Cattaneo
- Fondazione EBRI (European Brain Research Institute) Rita Levi-Montalcini, Rome, Italy.,BIO@SNS, Scuola Normale Superiore, Pisa, Italy
| |
Collapse
|
20
|
Landy MA, Goyal M, Casey KM, Liu C, Lai HC. Loss of Prdm12 during development, but not in mature nociceptors, causes defects in pain sensation. Cell Rep 2021; 34:108913. [PMID: 33789102 PMCID: PMC8048104 DOI: 10.1016/j.celrep.2021.108913] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/11/2021] [Accepted: 03/05/2021] [Indexed: 12/30/2022] Open
Abstract
Prdm12 is a key transcription factor in nociceptor neurogenesis. Mutations of Prdm12 cause congenital insensitivity to pain (CIP) from failure of nociceptor development. However, precisely how deletion of Prdm12 during development or adulthood affects nociception is unknown. Here, we employ tissue- and temporal-specific knockout mouse models to test the function of Prdm12 during development and in adulthood. We find that constitutive loss of Prdm12 causes deficiencies in proliferation during sensory neurogenesis. We also demonstrate that conditional knockout from dorsal root ganglia (DRGs) during embryogenesis causes defects in nociception. In contrast, we find that, in adult DRGs, Prdm12 is dispensable for most pain-sensation and injury-induced hypersensitivity. Using transcriptomic analysis, we find mostly unique changes in adult Prdm12 knockout DRGs compared with embryonic knockout and that PRDM12 is likely a transcriptional activator in the adult. Overall, we find that the function of PRDM12 changes over developmental time.
Collapse
Affiliation(s)
- Mark A Landy
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Megan Goyal
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Katherine M Casey
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chen Liu
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, Hypothalamic Research Center, Dallas, TX 75390, USA
| | - Helen C Lai
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
21
|
Brandi R, Fabiano M, Giorgi C, Arisi I, La Regina F, Malerba F, Turturro S, Storti AE, Ricevuti F, Amadio S, Volontè C, Capsoni S, Scardigli R, D’Onofrio M, Cattaneo A. Nerve Growth Factor Neutralization Promotes Oligodendrogenesis by Increasing miR-219a-5p Levels. Cells 2021; 10:cells10020405. [PMID: 33669304 PMCID: PMC7920049 DOI: 10.3390/cells10020405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 12/11/2022] Open
Abstract
In the brain, the neurotrophin Nerve growth factor (NGF) regulates not only neuronal survival and differentiation, but also glial and microglial functions and neuroinflammation. NGF is known to regulate oligodendrogenesis, reducing myelination in the central nervous system (CNS). In this study, we found that NGF controls oligodendrogenesis by modulating the levels of miR-219a-5p, a well-known positive regulator of oligodendrocyte differentiation. We exploited an NGF-deprivation mouse model, the AD11 mice, in which the postnatal expression of an anti-NGF antibody leads to NGF neutralization and progressive neurodegeneration. Notably, we found that these mice also display increased myelination. A microRNA profiling of AD11 brain samples and qRT-PCR analyses revealed that NGF deprivation leads to an increase of miR-219a-5p levels in hippocampus and cortex and a corresponding down-regulation of its predicted targets. Neurospheres isolated from the hippocampus of AD11 mice give rise to more oligodendrocytes and this process is dependent on miR-219a-5p, as shown by decoy-mediated inhibition of this microRNA. Moreover, treatment of AD11 neurospheres with NGF inhibits miR-219a-5p up-regulation and, consequently, oligodendrocyte differentiation, while anti-NGF treatment of wild type (WT) oligodendrocyte progenitors increases miR-219a-5p expression and the number of mature cells. Overall, this study indicates that NGF inhibits oligodendrogenesis and myelination by down-regulating miR-219a-5p levels, suggesting a novel molecular circuitry that can be exploited for the discovery of new effectors for remyelination in human demyelinating diseases, such as Multiple Sclerosis.
Collapse
Affiliation(s)
- Rossella Brandi
- European Brain Research Institute (EBRI) “Rita Levi-Montalcini”, Viale Regina Elena, 295, 00161 Rome, Italy; (R.B.); (M.F.); (C.G.); (I.A.); (F.L.R.); (F.M.); (S.T.); (A.E.S.); (F.R.)
| | - Marietta Fabiano
- European Brain Research Institute (EBRI) “Rita Levi-Montalcini”, Viale Regina Elena, 295, 00161 Rome, Italy; (R.B.); (M.F.); (C.G.); (I.A.); (F.L.R.); (F.M.); (S.T.); (A.E.S.); (F.R.)
| | - Corinna Giorgi
- European Brain Research Institute (EBRI) “Rita Levi-Montalcini”, Viale Regina Elena, 295, 00161 Rome, Italy; (R.B.); (M.F.); (C.G.); (I.A.); (F.L.R.); (F.M.); (S.T.); (A.E.S.); (F.R.)
- CNR, Institute of Molecular Biology and Pathology (IBPM), P.le Aldo Moro, 5, 00185 Rome, Italy
| | - Ivan Arisi
- European Brain Research Institute (EBRI) “Rita Levi-Montalcini”, Viale Regina Elena, 295, 00161 Rome, Italy; (R.B.); (M.F.); (C.G.); (I.A.); (F.L.R.); (F.M.); (S.T.); (A.E.S.); (F.R.)
- CNR, Institute of Translational Pharmacology (IFT), Via del Fosso del Cavaliere 100, 00131 Rome, Italy
| | - Federico La Regina
- European Brain Research Institute (EBRI) “Rita Levi-Montalcini”, Viale Regina Elena, 295, 00161 Rome, Italy; (R.B.); (M.F.); (C.G.); (I.A.); (F.L.R.); (F.M.); (S.T.); (A.E.S.); (F.R.)
| | - Francesca Malerba
- European Brain Research Institute (EBRI) “Rita Levi-Montalcini”, Viale Regina Elena, 295, 00161 Rome, Italy; (R.B.); (M.F.); (C.G.); (I.A.); (F.L.R.); (F.M.); (S.T.); (A.E.S.); (F.R.)
| | - Sabrina Turturro
- European Brain Research Institute (EBRI) “Rita Levi-Montalcini”, Viale Regina Elena, 295, 00161 Rome, Italy; (R.B.); (M.F.); (C.G.); (I.A.); (F.L.R.); (F.M.); (S.T.); (A.E.S.); (F.R.)
| | - Andrea Ennio Storti
- European Brain Research Institute (EBRI) “Rita Levi-Montalcini”, Viale Regina Elena, 295, 00161 Rome, Italy; (R.B.); (M.F.); (C.G.); (I.A.); (F.L.R.); (F.M.); (S.T.); (A.E.S.); (F.R.)
| | - Flavia Ricevuti
- European Brain Research Institute (EBRI) “Rita Levi-Montalcini”, Viale Regina Elena, 295, 00161 Rome, Italy; (R.B.); (M.F.); (C.G.); (I.A.); (F.L.R.); (F.M.); (S.T.); (A.E.S.); (F.R.)
| | - Susanna Amadio
- IRCCS Fondazione Santa Lucia, Preclinical Neuroscience, Via del Fosso di Fiorano 65, 00143 Rome, Italy; (S.A.); (C.V.)
| | - Cinzia Volontè
- IRCCS Fondazione Santa Lucia, Preclinical Neuroscience, Via del Fosso di Fiorano 65, 00143 Rome, Italy; (S.A.); (C.V.)
- CNR, Institute for Systems Analysis and Computer Science, Via Dei Taurini 19, 00185 Rome, Italy
| | - Simona Capsoni
- Bio@SNS, Scuola Normale Superiore, 56124 Pisa, Italy;
- Institute of Physiology, Department of Neuroscience and Rehabilitation University of Ferrara, 44121 Ferrara, Italy
| | - Raffaella Scardigli
- European Brain Research Institute (EBRI) “Rita Levi-Montalcini”, Viale Regina Elena, 295, 00161 Rome, Italy; (R.B.); (M.F.); (C.G.); (I.A.); (F.L.R.); (F.M.); (S.T.); (A.E.S.); (F.R.)
- CNR, Institute of Translational Pharmacology (IFT), Via del Fosso del Cavaliere 100, 00131 Rome, Italy
- Correspondence: (R.S.); (M.D.); (A.C.)
| | - Mara D’Onofrio
- European Brain Research Institute (EBRI) “Rita Levi-Montalcini”, Viale Regina Elena, 295, 00161 Rome, Italy; (R.B.); (M.F.); (C.G.); (I.A.); (F.L.R.); (F.M.); (S.T.); (A.E.S.); (F.R.)
- CNR, Institute of Translational Pharmacology (IFT), Via del Fosso del Cavaliere 100, 00131 Rome, Italy
- Correspondence: (R.S.); (M.D.); (A.C.)
| | - Antonino Cattaneo
- Bio@SNS, Scuola Normale Superiore, 56124 Pisa, Italy;
- Correspondence: (R.S.); (M.D.); (A.C.)
| |
Collapse
|
22
|
Abstract
Primary nociceptors are a heterogeneous class of peripheral somatosensory neurons, responsible for detecting noxious, pruriceptive, and thermal stimuli. These neurons are further divided into several molecularly defined subtypes that correlate with their functional sensory modalities and morphological features. During development, all nociceptors arise from a common pool of embryonic precursors, and then segregate progressively into their mature specialized phenotypes. In this review, we summarize the intrinsic transcriptional programs and extrinsic trophic factor signaling mechanisms that interact to control nociceptor diversification. We also discuss how recent transcriptome profiling studies have significantly advanced the field of sensory neuron development.
Collapse
Affiliation(s)
- Suna L Cranfill
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wenqin Luo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
23
|
Yang W, Sung K, Xu W, Rodriguez MJ, Wu AC, Santos SA, Fang S, Uber RK, Dong SX, Guillory BC, Orain X, Raus J, Jolivalt C, Calcutt N, Rissman RA, Ding J, Wu C. A missense point mutation in nerve growth factor (NGF R100W) results in selective peripheral sensory neuropathy. Prog Neurobiol 2020; 194:101886. [PMID: 32693191 DOI: 10.1016/j.pneurobio.2020.101886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 06/09/2020] [Accepted: 07/11/2020] [Indexed: 01/15/2023]
Abstract
The R100W mutation in nerve growth factor is associated with hereditary sensory autonomic neuropathy V in a Swedish family. These patients develop severe loss of perception to deep pain but with apparently normal cognitive functions. To better understand the disease mechanism, we examined a knockin mouse model of HSAN V. The homozygous mice showed significant structural deficits in intra-epidermal nerve fibers (IENFs) at birth. These mice had a total loss of pain perception at ∼2 months of age and often failed to survive to adulthood. Heterozygous mutant mice developed a progressive degeneration of small sensory fibers both behaviorally and functionally: they showed a progressive loss of IENFs starting at the age of 9 months accompanied with progressive loss of perception to painful stimuli such as noxious temperature. Quantitative analysis of lumbar 4/5 dorsal root ganglia revealed a significant reduction in small size neurons, while analysis of sciatic nerve fibers revealed the heterozygous mutant mice had no reduction in myelinated nerve fibers. Significantly, the amount of NGF secreted from mouse embryonic fibroblasts were reduced from both heterozygous and homozygous mice compared to their wild-type littermates. Interestingly, the heterozygous mice showed no apparent structural alteration in the brain: neither the anterior cingulate cortex nor the medial septum including NGF-dependent basal forebrain cholinergic neurons. Accordingly, these animals did not develop appreciable deficits in tests for brain function. Our study has thus demonstrated that the NGFR100W mutation likely affects the structure and function of peripheral sensory neurons.
Collapse
Affiliation(s)
- Wanlin Yang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; Department of Neurology, Zhuijiang Hospital, Southern Medical University, Guangzhou, China
| | - Kijung Sung
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Wei Xu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Maria J Rodriguez
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Andrew C Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Sarai A Santos
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Savannah Fang
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Rebecca K Uber
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Stephanie X Dong
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Brandon C Guillory
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Xavier Orain
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Jordan Raus
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Corrine Jolivalt
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Nigel Calcutt
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Jianqing Ding
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
24
|
Abstract
Supplemental Digital Content is Available in the Text. A ligand-guided, light-activated photosensitizer tool targets TrkA-expressing nociceptors, reversing acute and chronic pain in mice. Nerve growth factor (NGF) and its receptors TrkA and p75 play a key role in the development and function of peripheral nociceptive neurons. Here, we describe novel technology to selectively photoablate TrkA-positive nociceptors through delivery of a phototoxic agent coupled to an engineered NGF ligand and subsequent near-infrared illumination. We demonstrate that this approach allows for on demand and localized reversal of pain behaviors in mouse models of acute, inflammatory, neuropathic, and joint pain. To target peripheral nociceptors, we generated a SNAP-tagged NGF derivative NGFR121W that binds to TrkA/p75 receptors but does not provoke signaling in TrkA-positive cells or elicit pain behaviors in mice. NGFR121W-SNAP was coupled to the photosensitizer IRDye700DX phthalocyanine (IR700) and injected subcutaneously. After near-infrared illumination of the injected area, behavioral responses to nociceptive mechanical and sustained thermal stimuli, but not innocuous stimuli, were substantially reduced. Similarly, in models of inflammatory, osteoarthritic, and neuropathic pain, mechanical hypersensitivity was abolished for 3 weeks after a single treatment regime. We demonstrate that this loss of pain behavior coincides with the retraction of neurons from the skin which then reinnervate the epidermis after 3 weeks corresponding with the return of mechanical hypersensitivity. Thus NGFR121W-SNAP-mediated photoablation is a minimally invasive approach to reversibly silence nociceptor input from the periphery, and control pain and hypersensitivity to mechanical stimuli.
Collapse
|
25
|
Unveiling functional motions based on point mutations in biased signaling systems: A normal mode study on nerve growth factor bound to TrkA. PLoS One 2020; 15:e0231542. [PMID: 32497034 PMCID: PMC7272051 DOI: 10.1371/journal.pone.0231542] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 03/26/2020] [Indexed: 11/19/2022] Open
Abstract
Many receptors elicit signal transduction by activating multiple intracellular pathways. This transduction can be triggered by a non-specific ligand, which simultaneously activates all the signaling pathways of the receptors. However, the binding of one biased ligand preferentially trigger one pathway over another, in a process called biased signaling. The identification the functional motions related to each of these distinct pathways has a direct impact on the development of new effective and specific drugs. We show here how to detect specific functional motions by considering the case of the NGF/TrkA-Ig2 complex. NGF-mediated TrkA receptor activation is dependent on specific structural motions that trigger the neuronal growth, development, and survival of neurons in nervous system. The R221W mutation in the ngf gene impairs nociceptive signaling. We discuss how the large-scale structural effects of this mutation lead to the suppression of collective motions necessary to induce TrkA activation of nociceptive signaling. Our results suggest that subtle changes in the NGF interaction network due to the point mutation are sufficient to inhibit the motions of TrkA receptors putatively linked to nociception. The methodological approach presented in this article, based jointly on the normal mode analysis and the experimentally observed functional alterations due to point mutations provides an essential tool to reveal the structural changes and motions linked to the disease, which in turn could be necessary for a drug design study.
Collapse
|
26
|
Barker PA, Mantyh P, Arendt-Nielsen L, Viktrup L, Tive L. Nerve Growth Factor Signaling and Its Contribution to Pain. J Pain Res 2020; 13:1223-1241. [PMID: 32547184 PMCID: PMC7266393 DOI: 10.2147/jpr.s247472] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Nerve growth factor (NGF) is a neurotrophic protein essential for the growth, differentiation, and survival of sympathetic and sensory afferent neurons during development. A substantial body of evidence, based on both animal and human studies, demonstrates that NGF plays a pivotal role in modulation of nociception in adulthood. This has spurred development of a variety of novel analgesics that target the NGF signaling pathway. Here, we present a narrative review designed to summarize how NGF receptor activation and downstream signaling alters nociception through direct sensitization of nociceptors at the site of injury and changes in gene expression in the dorsal root ganglion that collectively increase nociceptive signaling from the periphery to the central nervous system. This review illustrates that NGF has a well-known and multifunctional role in nociceptive processing, although the precise signaling pathways downstream of NGF receptor activation that mediate nociception are complex and not completely understood. Additionally, much of the existing knowledge derives from studies performed in animal models and may not accurately represent the human condition. However, available data establish a role for NGF in the modulation of nociception through effects on the release of inflammatory mediators, nociceptive ion channel/receptor activity, nociceptive gene expression, and local neuronal sprouting. The role of NGF in nociception and the generation and/or maintenance of chronic pain has led to it becoming a novel and attractive target of pain therapeutics for the treatment of chronic pain conditions.
Collapse
Affiliation(s)
- Philip A Barker
- Department of Biology, University of British Columbia, Kelowna, BC, Canada
| | - Patrick Mantyh
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Lars Arendt-Nielsen
- Department of Health Science and Technology and the Center for Sensory-Motor Interaction/Center for Neuroplasticity and Pain, Aalborg University, Aalborg, Denmark
| | | | | |
Collapse
|
27
|
The NGF R100W Mutation Specifically Impairs Nociception without Affecting Cognitive Performance in a Mouse Model of Hereditary Sensory and Autonomic Neuropathy Type V. J Neurosci 2019; 39:9702-9715. [PMID: 31685654 DOI: 10.1523/jneurosci.0688-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 10/01/2019] [Accepted: 10/27/2019] [Indexed: 12/22/2022] Open
Abstract
Nerve growth factor (NGF) is a key mediator of nociception, acting during the development and differentiation of dorsal root ganglion (DRG) neurons, and on adult DRG neuron sensitization to painful stimuli. NGF also has central actions in the brain, where it regulates the phenotypic maintenance of cholinergic neurons. The physiological function of NGF as a pain mediator is altered in patients with Hereditary Sensory and Autonomic Neuropathy type V (HSAN V), caused by the 661C>T transition in the Ngf gene, resulting in the R100W missense mutation in mature NGF. Homozygous HSAN V patients present with congenital pain insensitivity, but are cognitively normal. This led us to hypothesize that the R100W mutation may differentially affect the central and peripheral actions of NGF. To test this hypothesis and provide a mechanistic basis to the HSAN V phenotype, we generated transgenic mice harboring the human 661C>T mutation in the Ngf gene and studied both males and females. We demonstrate that heterozygous NGFR100W/wt mice display impaired nociception. DRG neurons of NGFR100W/wt mice are morphologically normal, with no alteration in the different DRG subpopulations, whereas skin innervation is reduced. The NGFR100W protein has reduced capability to activate pain-specific signaling, paralleling its reduced ability to induce mechanical allodynia. Surprisingly, however, NGFR100W/wt mice, unlike heterozygous mNGF+/- mice, show no learning or memory deficits, despite a reduction in secretion and brain levels of NGF. The results exclude haploinsufficiency of NGF as a mechanistic cause for heterozygous HSAN V mice and demonstrate a specific effect of the R100W mutation on nociception.SIGNIFICANCE STATEMENT The R100W mutation in nerve growth factor (NGF) causes Hereditary Sensory and Autonomic Neuropathy type V, a rare disease characterized by impaired nociception, even in apparently clinically silent heterozygotes. For the first time, we generated and characterized heterozygous knock-in mice carrying the human R100W-mutated allele (NGFR100W/wt). Mutant mice have normal nociceptor populations, which, however, display decreased activation of pain transduction pathways. NGFR100W interferes with peripheral and central NGF bioavailability, but this does not impact on CNS function, as demonstrated by normal learning and memory, in contrast with heterozygous NGF knock-out mice. Thus, a point mutation allows neurotrophic and pronociceptive functions of NGF to be split, with interesting implications for the treatment of chronic pain.
Collapse
|
28
|
Dahlström M, Nordvall G, Sundström E, Åkesson E, Tegerstedt G, Eriksdotter M, Forsell P. Identification of amino acid residues of nerve growth factor important for neurite outgrowth in human dorsal root ganglion neurons. Eur J Neurosci 2019; 50:3487-3501. [PMID: 31301255 PMCID: PMC6899756 DOI: 10.1111/ejn.14513] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 06/27/2019] [Accepted: 07/05/2019] [Indexed: 01/21/2023]
Abstract
Nerve growth factor (NGF) is an essential neurotrophic factor for the development and maintenance of the central and the peripheral nervous system. NGF deficiency in the basal forebrain precedes degeneration of basal forebrain cholinergic neurons in Alzheimer's disease, contributing to memory decline. NGF mediates neurotrophic support via its high‐affinity receptor, the tropomyosin‐related kinase A (TrkA) receptor, and mediates mitogenic and differentiation signals via the extracellular signal‐regulated protein kinases 1 and 2 (ERK1/2). However, the molecular mechanisms underlying the different NGF/TrkA/ERK signalling pathways are far from clear. In this study, we have investigated the role of human NGF and three NGF mutants, R100E, W99A and K95A/Q96A, their ability to activate TrkA or ERK1/2, and their ability to induce proliferation or differentiation in human foetal dorsal root ganglion (DRG) neurons or in PC12 cells. We show that the R100E mutant was significantly more potent than NGF itself to induce proliferation and differentiation, and significantly more potent in activation of ERK1/2 in DRG neurons. The W99A and K95A/Q96A mutants, on the other hand, were less effective than the wild‐type protein. An unexpected finding was the high efficacy of the K95A/Q96A mutant to activate TrkA and to induce differentiation of DRG neurons at elevated concentrations. These data demonstrate an NGF mutant with improved neurotrophic properties in primary human neuronal cells. The R100E mutant represents an interesting candidate for further drug development in Alzheimer's disease and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Märta Dahlström
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,AlzeCure Foundation, Huddinge, Sweden
| | - Gunnar Nordvall
- AlzeCure Foundation, Huddinge, Sweden.,Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,AlzeCure Pharma AB, Huddinge, Sweden
| | - Erik Sundström
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Elisabet Åkesson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,R&D Unit, Stockholms Sjukhem, Stockholm, Sweden
| | - Gunilla Tegerstedt
- Division of Gynecology and Obstetrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Pontus Forsell
- AlzeCure Foundation, Huddinge, Sweden.,Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,AlzeCure Pharma AB, Huddinge, Sweden
| |
Collapse
|
29
|
Kaliyaperumal S, Wilson K, Aeffner F, Dean C. Animal Models of Peripheral Pain: Biology Review and Application for Drug Discovery. Toxicol Pathol 2019; 48:202-219. [PMID: 31269874 DOI: 10.1177/0192623319857051] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pain is a complex constellation of cognitive, unpleasant sensory, and emotional experiences that primarily serves as a survival mechanism. Pain arises in the peripheral nervous system and pain signals synapse with nerve tracts extending into the central nervous system. Several different schemes are used to classify pain, including the underlying mechanism, tissues primarily affected, and time-course. Numerous animal models of pain, which should be employed with appropriate Institutional Animal Care and Use approvals, have been developed to elucidate pathophysiology mechanisms and aid in identification of novel therapeutic targets. The variety of available models underscores the observations that pain phenotypes are driven by several distinct mechanisms. Pain outcome measurement encompasses both reflexive (responses to heat, cold, mechanical and electrical stimuli) and nonreflexive (spontaneous pain responses to stimuli) behaviors. However, the question of translatability to human pain conditions and potential treatment outcomes remains a topic of continued scrutiny. In this review we discuss the different types of pain and their mechanisms and pathways, available rodent pain models with an emphasis on type of pain stimulations and pain outcome measures and discuss the role of pathologists in assessing and validating pain models.
Collapse
Affiliation(s)
| | | | | | - Charles Dean
- Amgen, Inc, Thousand Oaks, CA, USA *Both authors equally contributed to the manuscript
| |
Collapse
|
30
|
Haploinsufficiency of the brain-derived neurotrophic factor gene is associated with reduced pain sensitivity. Pain 2019; 160:1070-1081. [PMID: 30855519 DOI: 10.1097/j.pain.0000000000001485] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Rare pain-insensitive individuals offer unique insights into how pain circuits function and have led to the development of new strategies for pain control. We investigated pain sensitivity in humans with WAGR (Wilms tumor, aniridia, genitourinary anomaly, and range of intellectual disabilities) syndrome, who have variably sized heterozygous deletion of the 11p13 region. The deletion region can be inclusive or exclusive of the brain-derived neurotrophic factor (BDNF) gene, a crucial trophic factor for nociceptive afferents. Nociceptive responses assessed by quantitative sensory testing demonstrated reduced pain sensitivity only in the WAGR subjects whose deletion boundaries included the BDNF gene. Corresponding behavioral assessments were made in heterozygous Bdnf knockout rats to examine the specific role of Bdnf. These analogous experiments revealed impairment of Aδ- and C-fiber-mediated heat nociception, determined by acute nociceptive thermal stimuli, and in aversive behaviors evoked when the rats were placed on a hot plate. Similar results were obtained for C-fiber-mediated cold responses and cold avoidance on a cold-plate device. Together, these results suggested a blunted responsiveness to aversive stimuli. Our parallel observations in humans and rats show that hemizygous deletion of the BDNF gene reduces pain sensitivity and establishes BDNF as a determinant of nociceptive sensitivity.
Collapse
|
31
|
Sheehan K, Lee J, Chong J, Zavala K, Sharma M, Philipsen S, Maruyama T, Xu Z, Guan Z, Eilers H, Kawamata T, Schumacher M. Transcription factor Sp4 is required for hyperalgesic state persistence. PLoS One 2019; 14:e0211349. [PMID: 30811405 PMCID: PMC6392229 DOI: 10.1371/journal.pone.0211349] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/11/2019] [Indexed: 12/14/2022] Open
Abstract
Understanding how painful hypersensitive states develop and persist beyond the initial hours to days is critically important in the effort to devise strategies to prevent and/or reverse chronic painful states. Changes in nociceptor transcription can alter the abundance of nociceptive signaling elements, resulting in longer-term change in nociceptor phenotype. As a result, sensitized nociceptive signaling can be further amplified and nocifensive behaviors sustained for weeks to months. Building on our previous finding that transcription factor Sp4 positively regulates the expression of the pain transducing channel TRPV1 in Dorsal Root Ganglion (DRG) neurons, we sought to determine if Sp4 serves a broader role in the development and persistence of hypersensitive states in mice. We observed that more than 90% of Sp4 staining DRG neurons were small to medium sized, primarily unmyelinated (NF200 neg) and the majority co-expressed nociceptor markers TRPV1 and/or isolectin B4 (IB4). Genetically modified mice (Sp4+/-) with a 50% reduction of Sp4 showed a reduction in DRG TRPV1 mRNA and neuronal responses to the TRPV1 agonist-capsaicin. Importantly, Sp4+/- mice failed to develop persistent inflammatory thermal hyperalgesia, showing a reversal to control values after 6 hours. Despite a reversal of inflammatory thermal hyperalgesia, there was no difference in CFA-induced hindpaw swelling between CFA Sp4+/- and CFA wild type mice. Similarly, Sp4+/- mice failed to develop persistent mechanical hypersensitivity to hind-paw injection of NGF. Although Sp4+/- mice developed hypersensitivity to traumatic nerve injury, Sp4+/- mice failed to develop persistent cold or mechanical hypersensitivity to the platinum-based chemotherapeutic agent oxaliplatin, a non-traumatic model of neuropathic pain. Overall, Sp4+/- mice displayed a remarkable ability to reverse the development of multiple models of persistent inflammatory and neuropathic hypersensitivity. This suggests that Sp4 functions as a critical control point for a network of genes that conspire in the persistence of painful hypersensitive states.
Collapse
Affiliation(s)
- Kayla Sheehan
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Jessica Lee
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Jillian Chong
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Kathryn Zavala
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Manohar Sharma
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Sjaak Philipsen
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tomoyuki Maruyama
- Department of Anesthesiology, Wakayama Medical University, Wakayama, Japan
| | - Zheyun Xu
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Helge Eilers
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
| | - Tomoyuki Kawamata
- Department of Anesthesiology, Wakayama Medical University, Wakayama, Japan
| | - Mark Schumacher
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
32
|
Sung K, Yang W, Wu C. Uncoupling neurotrophic function from nociception of nerve growth factor: what can be learned from a rare human disease? Neural Regen Res 2019; 14:570-573. [PMID: 30632491 PMCID: PMC6352596 DOI: 10.4103/1673-5374.247442] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Nerve growth factor (NGF) is a powerful trophic factor that provides essential support for the survival and differentiation of sympathetic and sensory neurons during development. However, NGF also activates nociceptors contributing significantly to inflammatory pain and neuropathic pain after tissue injury. As such anti-NGF based therapies represent a promising strategy for pain management. Because of dose-dependent serious side effects such as back pain, injection site hyperalgesia, clinical trials of using NGF to treat various disorders such as diabetic neuropathies, chemotherapy-induced and human immunodeficiency virus-associated peripheral neuropathies were all discontinued. Thus far, worldwide clinical applications of NGF in treating patients are very limited except in China. Hereditary sensory autonomic neuropathy type V (HSAN V) is an extremely rare disease. Genetic analyses have revealed that HSAN V is associated with autosomal recessive mutations in NGF. One of the mutations occurred at the 100th position of mature NGF resulting in a change of residue from arginine to tryptophan (R100W). Although those HSAN V patients associated with the NGFR100W mutation suffer from severe loss of deep pain, bone fractures and joint destruction, interestingly patients with the NGFR100W mutation do not show apparent cognitive deficits, suggesting important trophic support function is preserved. We believe that NGFR100W provides an ideal tool to uncouple the two important functions of NGF: trophic versus nociceptive. Studies from investigators including ourselves have indeed confirmed in animal testing that the NGFR100W no longer induced pain. More importantly, the trophic function seemed to be largely preserved in NGF harboring the R100W mutation. On the mechanistic level, we found that the NGFR100W mutation was capable of binding to and signaling through the tyrosine receptor kinase A receptor. But its ability to bind to and activate the 75 kDa neurotrophic factor was significantly diminished. The significance of these findings is at least two folds: 1) the NGFR100W mutation can be used as an alternative to the wildtype NGF to treat human conditions without eliciting pain; and 2) the 75 kDa neurotrophic factor may serve as a novel target for pain management. We will discuss all the details in this mini-review.
Collapse
Affiliation(s)
- Kijung Sung
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Wanlin Yang
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
33
|
Yang W, Sung K, Zhou F, Xu W, Rissman RA, Ding J, Wu C. Targeted Mutation (R100W) of the Gene Encoding NGF Leads to Deficits in the Peripheral Sensory Nervous System. Front Aging Neurosci 2018; 10:373. [PMID: 30524266 PMCID: PMC6262302 DOI: 10.3389/fnagi.2018.00373] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 10/26/2018] [Indexed: 12/25/2022] Open
Abstract
Nerve growth factor (NGF) exerts multifaceted functions through different stages of life. A missense mutation (R100W) in the beta-NGF gene was found in hereditary sensory autonomic neuropathy V (HSAN V) patients with severe loss of pain perception but without overt cognitive impairment. To better understand the pathogenesis of HSAN V, we generated the first NGFR100W knock in mouse model for HSAN V. We found that the homozygotes exhibited a postnatal lethal phenotype. A majority of homozygous pups died within the first week. Some homozygous pups could ingest more milk and survived up to 2 months by reducing litter size. Whole mount in situ hybridization using E10.5 embryos revealed that, compared to wild type, R100W mutation did not alter the gene expression patterns of TrkA and P75NTR in the homozygotes. We also found that the homozygotes displayed normal embryonic development of major organs (heart, lung, liver, kidney, and spleen). Furthermore, the homozygotes exhibited severe loss of PGP9.5-positive intra-epidermal sensory fibers. Taken together, our results suggest that, as with HSAN V patients, the R100W mutation primarily affects the peripheral sensory nervous system in the mouse model. This novel mouse model makes it possible to further study in vivo how NGFR100W uncouple trophic function from nociception of NGF.
Collapse
Affiliation(s)
- Wanlin Yang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kijung Sung
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Fengli Zhou
- Department of Respiratory Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Xu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Robert A. Rissman
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
- Veterans Affairs San Diego Health Care System, San Diego, CA, United States
| | - Jianqing Ding
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengbiao Wu
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
- Veterans Affairs San Diego Health Care System, San Diego, CA, United States
| |
Collapse
|
34
|
Cattaneo A, Capsoni S. Painless Nerve Growth Factor: A TrkA biased agonist mediating a broad neuroprotection via its actions on microglia cells. Pharmacol Res 2018; 139:17-25. [PMID: 30391352 DOI: 10.1016/j.phrs.2018.10.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/28/2018] [Accepted: 10/28/2018] [Indexed: 12/13/2022]
Abstract
Nerve Growth Factor (NGF) is a therapeutic candidate for Alzheimer's disease, based on its well known actions on basal forebrain cholinergic neurons. However, because of its pro-nociceptive activity, in current clinical trials NGF has to be administered intraparenchymally into the brain by neurosurgery via cell or gene therapy approaches. To prevent the NGF pain-inducing collateral effects, thus avoiding the necessity for local brain injection, we developed painless NGF (hNGFp), based on the human genetic disease Hereditary Sensory and Autonomic Neuropathy type V (HSAN V). hNGFp has similar neurotrophic activity as wild type human NGF, but its pain sensitizing activity is tenfold lower. Pharmacologically, hNGFp is a biased receptor agonist of NGF TrkA receptor. The results of recent studies shed new light on the neuroprotective mechanism by hNGFp and are highly relevant for the planning of NGF-based clinical trials. The intraparenchymal delivery of hNGFp, as used in clinical trials, was simulated in the 5xFAD mouse model and found to be inefficacious in reducing Aβ plaque load. On the contrary, the same dose of hNGFp administered intranasally, which was rather widely biodistributed in the brain and did not induce pain sensitization, blocked APP processing into amyloid and restored synaptic plasticity and memory in this aggressive neurodegeneration model. This potent and broad neuroprotection by hNGFp was found to be mediated by hNGFp actions on glial cells. hNGFp increases inflammatory proteins such as the soluble TNFα receptor II and the chemokine CXCL12. Independent work has shown that NGF has a potent anti-inflammatory action on microglia and steers them towards a neuroprotective phenotype. These studies demonstrate that microglia cells are a new target cell of NGF in the brain and have therapeutic significance: i) they establish that the neuroprotective actions of hNGFp relies on a widespread exposure of the brain, ii) they identify a new anti-neurodegenerative pathway, linking hNGFp to inflammatory chemokines and cytokines via microglia, a common target for new therapeutic opportunities for neurodegenerative diseases, iii) they extend the neuroprotective potential of hNGFp beyond its classical cholinergic target, thereby widening the range of neurological diseases for which this neurotrophic factor might be used therapeutically, iv) they help interpreting the results of current NGF clinical trials in AD and the design of future trials with this new potent therapeutic candidate.
Collapse
Affiliation(s)
- Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy; Rita Levi-Montalcini European Brain Research Institute (EBRI), Roma, Italy.
| | - Simona Capsoni
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy; Section of Human Physiology, Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
35
|
Sung K, Ferrari LF, Yang W, Chung C, Zhao X, Gu Y, Lin S, Zhang K, Cui B, Pearn ML, Maloney MT, Mobley WC, Levine JD, Wu C. Swedish Nerve Growth Factor Mutation (NGF R100W) Defines a Role for TrkA and p75 NTR in Nociception. J Neurosci 2018; 38:3394-3413. [PMID: 29483280 PMCID: PMC5895035 DOI: 10.1523/jneurosci.1686-17.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 01/23/2018] [Accepted: 02/12/2018] [Indexed: 12/14/2022] Open
Abstract
Nerve growth factor (NGF) exerts multiple functions on target neurons throughout development. The recent discovery of a point mutation leading to a change from arginine to tryptophan at residue 100 in the mature NGFβ sequence (NGFR100W) in patients with hereditary sensory and autonomic neuropathy type V (HSAN V) made it possible to distinguish the signaling mechanisms that lead to two functionally different outcomes of NGF: trophic versus nociceptive. We performed extensive biochemical, cellular, and live-imaging experiments to examine the binding and signaling properties of NGFR100W Our results show that, similar to the wild-type NGF (wtNGF), the naturally occurring NGFR100W mutant was capable of binding to and activating the TrkA receptor and its downstream signaling pathways to support neuronal survival and differentiation. However, NGFR100W failed to bind and stimulate the 75 kDa neurotrophic factor receptor (p75NTR)-mediated signaling cascades (i.e., the RhoA-Cofilin pathway). Intraplantar injection of NGFR100W into adult rats induced neither TrkA-mediated thermal nor mechanical acute hyperalgesia, but retained the ability to induce chronic hyperalgesia based on agonism for TrkA signaling. Together, our studies provide evidence that NGFR100W retains trophic support capability through TrkA and one aspect of its nociceptive signaling, but fails to engage p75NTR signaling pathways. Our findings suggest that wtNGF acts via TrkA to regulate the delayed priming of nociceptive responses. The integration of both TrkA and p75NTR signaling thus appears to regulate neuroplastic effects of NGF in peripheral nociception.SIGNIFICANCE STATEMENT In the present study, we characterized the naturally occurring nerve growth factor NGFR100W mutant that is associated with hereditary sensory and autonomic neuropathy type V. We have demonstrated for the first time that NGFR100W retains trophic support capability through TrkA, but fails to engage p75NTR signaling pathways. Furthermore, after intraplantar injection into adult rats, NGFR100W induced neither thermal nor mechanical acute hyperalgesia, but retained the ability to induce chronic hyperalgesia. We have also provided evidence that the integration of both TrkA- and p75NTR-mediated signaling appears to regulate neuroplastic effects of NGF in peripheral nociception. Our study with NGFR100W suggests that it is possible to uncouple trophic effect from nociceptive function, both induced by wild-type NGF.
Collapse
Affiliation(s)
| | - Luiz F Ferrari
- Department of Oral Surgery, University of California San Francisco, San Francisco, California 94143
| | - Wanlin Yang
- Department of Neurosciences
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China 200025
| | - ChiHye Chung
- Department of Biological Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 143-701, South Korea
| | | | - Yingli Gu
- Department of Neurosciences
- Department of Neurology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China 150001
| | - Suzhen Lin
- Department of Neurosciences
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China 200025
| | - Kai Zhang
- Department of Chemistry
- Department of Biochemistry, Neuroscience Program, Center for Biophysics and Quantitative Biology, Chemistry-Biology Interface Training Program, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, and
| | | | - Matthew L Pearn
- Department of Anesthesiology, University of California San Diego, School of Medicine, La Jolla, California 92093
- V.A. San Diego Healthcare System, San Diego, California 92161
| | - Michael T Maloney
- Department of Neurosciences, Stanford University, Stanford, California 94305
| | | | - Jon D Levine
- Department of Oral Surgery, University of California San Francisco, San Francisco, California 94143
| | - Chengbiao Wu
- Department of Neurosciences,
- V.A. San Diego Healthcare System, San Diego, California 92161
| |
Collapse
|
36
|
Zorina-Lichtenwalter K, Parisien M, Diatchenko L. Genetic studies of human neuropathic pain conditions: a review. Pain 2018; 159:583-594. [PMID: 29240606 PMCID: PMC5828382 DOI: 10.1097/j.pain.0000000000001099] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 10/20/2017] [Accepted: 10/26/2017] [Indexed: 12/12/2022]
Abstract
Numerous studies have shown associations between genetic variants and neuropathic pain disorders. Rare monogenic disorders are caused by mutations of substantial effect size in a single gene, whereas common disorders are likely to have a contribution from multiple genetic variants of mild effect size, representing different biological pathways. In this review, we survey the reported genetic contributors to neuropathic pain and submit them for validation in a 150,000-participant sample of the U.K. Biobank cohort. Successfully replicated association with a neuropathic pain construct for 2 variants in IL10 underscores the importance of neuroimmune interactions, whereas genome-wide significant association with low back pain (P = 1.3e-8) and false discovery rate 5% significant associations with hip, knee, and neck pain for variant rs7734804 upstream of the MAT2B gene provide evidence of shared contributing mechanisms to overlapping pain conditions at the molecular genetic level.
Collapse
Affiliation(s)
| | - Marc Parisien
- Alan Edwards Pain Centre, McGill University, Montreal, QC, Canada
| | - Luda Diatchenko
- Alan Edwards Pain Centre, McGill University, Montreal, QC, Canada
| |
Collapse
|
37
|
Clathrin heavy chain 22 contributes to the control of neuropeptide degradation and secretion during neuronal development. Sci Rep 2018; 8:2340. [PMID: 29402896 PMCID: PMC5799199 DOI: 10.1038/s41598-018-19980-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 01/09/2018] [Indexed: 02/02/2023] Open
Abstract
The repertoire of cell types in the human nervous system arises through a highly orchestrated process, the complexity of which is still being discovered. Here, we present evidence that CHC22 has a non-redundant role in an early stage of neural precursor differentiation, providing a potential explanation of why CHC22 deficient patients are unable to feel touch or pain. We show the CHC22 effect on neural differentiation is independent of the more common clathrin heavy chain CHC17, and that CHC22-dependent differentiation is mediated through an autocrine/paracrine mechanism. Using quantitative proteomics, we define the composition of clathrin-coated vesicles in SH-SY5Y cells, and determine proteome changes induced by CHC22 depletion. In the absence of CHC22 a subset of dense core granule (DCG) neuropeptides accumulated, were processed into biologically active 'mature' forms, and secreted in sufficient quantity to trigger neural differentiation. When CHC22 is present, however, these DCG neuropeptides are directed to the lysosome and degraded, thus preventing differentiation. This suggests that the brief reduction seen in CHC22 expression in sensory neural precursors may license a step in neuron precursor neurodevelopment; and that this step is mediated through control of a novel neuropeptide processing pathway.
Collapse
|
38
|
Shaikh SS, Nahorski MS, Woods CG. A third HSAN5 mutation disrupts the nerve growth factor furin cleavage site. Mol Pain 2018; 14:1744806918809223. [PMID: 30296891 PMCID: PMC6207963 DOI: 10.1177/1744806918809223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/20/2018] [Accepted: 09/20/2018] [Indexed: 11/17/2022] Open
Abstract
Bi-allelic dysfunctional mutations in nerve growth factor (NGF) cause the rare human phenotype hereditary sensory and autonomic neuropathy type 5 (HSAN5). We describe a novel NGF mutation in an individual with typical HSAN5 findings. The mutation c.361C>T, p.R121W is at the last residue of the furin cleavage motif Arg-Ser-Lys-Arg in proNGF. We show that the p.R121W mutation completely abolishes the formation of mature NGF-β. Surprisingly, mutant p.R121W cells produced very little proNGF. Instead, the two progressive cleavage products of proNGF were produced, proA-NGF and proB-NGF, with proB-NGF being the predominant NGF-derived peptide and the only peptide secreted by mutant p.R121W cells. We found that the ability of the p.R121W mutation to cause tropomyosin receptor kinase A autophosphorylation and mitogen-activated protein kinase phosphorylation was significantly reduced compared to controls (p < 0.05 and p < 0.01). By studying the PC12 cell line morphology and neurite length over a week, we found the p.R121W mutation had residual, but much reduced, neurotrophic activity when compared to wild-type NGF. Finally, we assessed whether the p.R121W mutation affected apoptosis and found a reduced protective effect compared to wild-type NGF. Our results suggest that the p.R121W NGF mutation causes HSAN5 through negating the ability of furin to cleave proNGF to produce NGF-β.
Collapse
Affiliation(s)
- Samiha S Shaikh
- Cambridge Institute for Medical Research, Addenbrooke's Biomedical Research Centre, Cambridge, UK
| | - Michael S Nahorski
- Cambridge Institute for Medical Research, Addenbrooke's Biomedical Research Centre, Cambridge, UK
| | - C Geoffrey Woods
- Cambridge Institute for Medical Research, Addenbrooke's Biomedical Research Centre, Cambridge, UK
- Department of Clinical Genetics, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
39
|
Kennedy AE, Vohra R, Scott JA, Ross GM. Effects of serum albumin on SPR-measured affinity of small molecule inhibitors binding to nerve growth factor. SENSING AND BIO-SENSING RESEARCH 2017. [DOI: 10.1016/j.sbsr.2017.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
40
|
Sidorova YA, Bespalov MM, Wong AW, Kambur O, Jokinen V, Lilius TO, Suleymanova I, Karelson G, Rauhala PV, Karelson M, Osborne PB, Keast JR, Kalso EA, Saarma M. A Novel Small Molecule GDNF Receptor RET Agonist, BT13, Promotes Neurite Growth from Sensory Neurons in Vitro and Attenuates Experimental Neuropathy in the Rat. Front Pharmacol 2017; 8:365. [PMID: 28680400 PMCID: PMC5478727 DOI: 10.3389/fphar.2017.00365] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 05/26/2017] [Indexed: 12/25/2022] Open
Abstract
Neuropathic pain caused by nerve damage is a common and severe class of chronic pain. Disease-modifying clinical therapies are needed as current treatments typically provide only symptomatic relief; show varying clinical efficacy; and most have significant adverse effects. One approach is targeting either neurotrophic factors or their receptors that normalize sensory neuron function and stimulate regeneration after nerve damage. Two candidate targets are glial cell line-derived neurotrophic factor (GDNF) and artemin (ARTN), as these GDNF family ligands (GFLs) show efficacy in animal models of neuropathic pain (Boucher et al., 2000; Gardell et al., 2003; Wang et al., 2008, 2014). As these protein ligands have poor drug-like properties and are expensive to produce for clinical use, we screened 18,400 drug-like compounds to develop small molecules that act similarly to GFLs (GDNF mimetics). This screening identified BT13 as a compound that selectively targeted GFL receptor RET to activate downstream signaling cascades. BT13 was similar to NGF and ARTN in selectively promoting neurite outgrowth from the peptidergic class of adult sensory neurons in culture, but was opposite to ARTN in causing neurite elongation without affecting initiation. When administered after spinal nerve ligation in a rat model of neuropathic pain, 20 and 25 mg/kg of BT13 decreased mechanical hypersensitivity and normalized expression of sensory neuron markers in dorsal root ganglia. In control rats, BT13 had no effect on baseline mechanical or thermal sensitivity, motor coordination, or weight gain. Thus, small molecule BT13 selectively activates RET and offers opportunities for developing novel disease-modifying medications to treat neuropathic pain.
Collapse
Affiliation(s)
- Yulia A Sidorova
- Laboratory of Molecular Neuroscience, Institute of Biotechnology, University of HelsinkiHelsinki, Finland
| | - Maxim M Bespalov
- Laboratory of Molecular Neuroscience, Institute of Biotechnology, University of HelsinkiHelsinki, Finland
| | - Agnes W Wong
- Department of Anatomy and Neuroscience, The University of MelbourneMelbourne, VIC, Australia
| | - Oleg Kambur
- Department of Pharmacology, Faculty of Medicine, University of HelsinkiHelsinki, Finland
| | - Viljami Jokinen
- Department of Pharmacology, Faculty of Medicine, University of HelsinkiHelsinki, Finland
| | - Tuomas O Lilius
- Department of Pharmacology, Faculty of Medicine, University of HelsinkiHelsinki, Finland
| | - Ilida Suleymanova
- Laboratory of Molecular Neuroscience, Institute of Biotechnology, University of HelsinkiHelsinki, Finland
| | | | - Pekka V Rauhala
- Department of Pharmacology, Faculty of Medicine, University of HelsinkiHelsinki, Finland
| | - Mati Karelson
- Department of Molecular Technology, Institute of Chemistry, University of TartuTartu, Estonia
| | - Peregrine B Osborne
- Department of Anatomy and Neuroscience, The University of MelbourneMelbourne, VIC, Australia
| | - Janet R Keast
- Department of Anatomy and Neuroscience, The University of MelbourneMelbourne, VIC, Australia
| | - Eija A Kalso
- Department of Pharmacology, Faculty of Medicine, University of HelsinkiHelsinki, Finland.,Pain Clinic, Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University HospitalHelsinki, Finland
| | - Mart Saarma
- Laboratory of Molecular Neuroscience, Institute of Biotechnology, University of HelsinkiHelsinki, Finland
| |
Collapse
|
41
|
Nanotechnological strategies for nerve growth factor delivery: Therapeutic implications in Alzheimer’s disease. Pharmacol Res 2017; 120:68-87. [DOI: 10.1016/j.phrs.2017.03.020] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 02/23/2017] [Accepted: 03/22/2017] [Indexed: 12/30/2022]
|
42
|
Severini C, Petrocchi Passeri P, Ciotti M, Florenzano F, Petrella C, Malerba F, Bruni B, D'Onofrio M, Arisi I, Brandi R, Possenti R, Calissano P, Cattaneo A. Nerve growth factor derivative NGF61/100 promotes outgrowth of primary sensory neurons with reduced signs of nociceptive sensitization. Neuropharmacology 2017; 117:134-148. [DOI: 10.1016/j.neuropharm.2017.01.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 01/09/2017] [Accepted: 01/29/2017] [Indexed: 12/16/2022]
|
43
|
Single-Fiber Recordings of Nociceptive Fibers in Patients With HSAN Type V With Congenital Insensitivity to Pain. Clin J Pain 2016; 32:636-42. [DOI: 10.1097/ajp.0000000000000303] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
44
|
Perini I, Tavakoli M, Marshall A, Minde J, Morrison I. Rare human nerve growth factor-β mutation reveals relationship between C-afferent density and acute pain evaluation. J Neurophysiol 2016; 116:425-30. [PMID: 27146986 DOI: 10.1152/jn.00667.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 04/29/2016] [Indexed: 11/22/2022] Open
Abstract
The rare nerve growth factor-β (NGFB) mutation R221W causes a selective loss of thinly myelinated fibers and especially unmyelinated C-fibers. Carriers of this mutation show altered pain sensation. A subset presents with arthropathic symptoms, with the homozygous most severely affected. The aim of the present study was to investigate the relationship between peripheral afferent loss and pain evaluation by performing a quantification of small-fiber density in the cornea of the carriers, relating density to pain evaluation measures. In vivo corneal confocal microscopy (CCM) was used to quantify C-fiber loss in the cornea of 19 R221W mutation carriers (3 homozygous) and 19 age-matched healthy control subjects. Pain evaluation data via the Situational Pain Questionnaire (SPQ) and the severity of neuropathy based on the Neuropathy Disability Score (NDS) were assessed. Homozygotes, heterozygotes, and control groups differed significantly in corneal C-nerve fiber density, with the homozygotes showing a significant afferent reduction. Importantly, peripheral C-fiber loss correlated negatively with pain evaluation, as revealed by SPQ scores. This study is the first to investigate the contribution of small-fiber density to the perceptual evaluation of pain. It demonstrates that the lower the peripheral small-fiber density, the lower the degree of reported pain intensity, indicating a functional relationship between small-fiber density and higher level pain experience.
Collapse
Affiliation(s)
- Irene Perini
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden;
| | - Mitra Tavakoli
- Centre for Endocrinology and Diabetes, Institute of Human Development, University of Manchester, Manchester, United Kingdom; National Institute of Health Research Collaboration for Leadership in Applied Health Research and Care Greater Manchester, Manchester, United Kingdom; University of Exeter Medical School, Exeter, United Kingdom
| | - Andrew Marshall
- Greater Manchester Neuroscience Centre, Salford Royal Hospital NHS Trust, Salford, United Kingdom; University of Manchester, Manchester, United Kingdom; Liverpool John Moores University, Liverpool, United Kingdom; and
| | - Jan Minde
- Department of Surgery, Unit of Orthopedics, Perioperative Sciences, Umeå University Hospital, Umeå, Sweden
| | - India Morrison
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
45
|
Neurotrophic factors and their inhibitors in chronic pain treatment. Neurobiol Dis 2016; 97:127-138. [PMID: 27063668 DOI: 10.1016/j.nbd.2016.03.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/24/2016] [Accepted: 03/30/2016] [Indexed: 11/21/2022] Open
Abstract
Chronic pain affects more than 20% of the UK population. Neurotrophic factors have been identified as therapeutic targets to improve current treatments of chronic pain. This review article focuses on nerve growth factor (NGF) and interleukin-6 (IL-6) as potential therapeutic targets. In this review we highlight the mechanisms of action and the current progress of targeted therapies in clinical trials.
Collapse
|
46
|
Nahorski MS, Chen YC, Woods CG. New Mendelian Disorders of Painlessness. Trends Neurosci 2015; 38:712-724. [DOI: 10.1016/j.tins.2015.08.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 02/08/2023]
|
47
|
Malerba F, Paoletti F, Bruni Ercole B, Materazzi S, Nassini R, Coppi E, Patacchini R, Capsoni S, Lamba D, Cattaneo A. Functional Characterization of Human ProNGF and NGF Mutants: Identification of NGF P61SR100E as a "Painless" Lead Investigational Candidate for Therapeutic Applications. PLoS One 2015; 10:e0136425. [PMID: 26371475 PMCID: PMC4570711 DOI: 10.1371/journal.pone.0136425] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/04/2015] [Indexed: 11/21/2022] Open
Abstract
Background Nerve Growth Factor (NGF) holds a great therapeutic promise for Alzheimer's disease, diabetic neuropathies, ophthalmic diseases, dermatological ulcers. However, the necessity for systemic delivery has hampered the clinical applications of NGF due to its potent pro-nociceptive action. A “painless” human NGF (hNGF R100E) mutant has been engineered. It has equal neurotrophic potency to hNGF but a lower nociceptive activity. We previously described and characterized the neurotrophic and nociceptive properties also of the hNGF P61S and P61SR100E mutants, selectively detectable against wild type hNGF. However, the reduced pain-sensitizing potency of the “painless” hNGF mutants has not been quantified. Objectives and Results Aiming at the therapeutic application of the “painless” hNGF mutants, we report on the comparative functional characterization of the precursor and mature forms of the mutants hNGF R100E and hNGF P61SR100E as therapeutic candidates, also in comparison to wild type hNGF and to hNGF P61S. The mutants were assessed by a number of biochemical, biophysical methods and assayed by cellular assays. Moreover, a highly sensitive ELISA for the detection of the P61S-tagged mutants in biological samples has been developed. Finally, we explored the pro-nociceptive effects elicited by hNGF mutants in vivo, demonstrating an expanded therapeutic window with a ten-fold increase in potency. Conclusions This structure-activity relationship study has led to validate the concept of developing painless NGF as a therapeutic, targeting the NGF receptor system and supporting the choice of hNGF P61S R100E as the best candidate to advance in clinical development. Moreover, this study contributes to the identification of the molecular determinants modulating the properties of the hNGF “painless” mutants.
Collapse
Affiliation(s)
- Francesca Malerba
- Neurotrophic Factors and Neurodegenerative Diseases Unit, European Brain Research Institute, “Rita Levi-Montalcini” Foundation, Rome, Italy
- Neurobiology Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
| | - Francesca Paoletti
- Neurotrophic Factors and Neurodegenerative Diseases Unit, European Brain Research Institute, “Rita Levi-Montalcini” Foundation, Rome, Italy
- Neurobiology Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
| | - Bruno Bruni Ercole
- Neurotrophic Factors and Neurodegenerative Diseases Unit, European Brain Research Institute, “Rita Levi-Montalcini” Foundation, Rome, Italy
| | - Serena Materazzi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Romina Nassini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Elisabetta Coppi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | | | - Simona Capsoni
- Neurobiology Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
| | - Doriano Lamba
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Area Science Park–Basovizza, Trieste, Italy
| | - Antonino Cattaneo
- Neurotrophic Factors and Neurodegenerative Diseases Unit, European Brain Research Institute, “Rita Levi-Montalcini” Foundation, Rome, Italy
- Neurobiology Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
- * E-mail:
| |
Collapse
|
48
|
Travaglia A, Pietropaolo A, Di Martino R, Nicoletti VG, La Mendola D, Calissano P, Rizzarelli E. A small linear peptide encompassing the NGF N-terminus partly mimics the biological activities of the entire neurotrophin in PC12 cells. ACS Chem Neurosci 2015; 6:1379-92. [PMID: 25939060 DOI: 10.1021/acschemneuro.5b00069] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ever since the discovery of its neurite growth promoting activity in sympathetic and sensory ganglia, nerve growth factor (NGF) became the prototype of the large family of neurotrophins. The use of primary cultures and clonal cell lines has revealed several distinct actions of NGF and other neurotrophins. Among several models of NGF activity, the clonal cell line PC12 is the most widely employed. Thus, in the presence of NGF, through the activation of the transmembrane protein TrkA, these cells undergo a progressive mitotic arrest and start to grow electrically excitable neuritis. A vast number of studies opened intriguing aspects of NGF mechanisms of action, its biological properties, and potential use as therapeutic agents. In this context, identifying and utilizing small portions of NGF is of great interest and involves several human diseases including Alzheimer's disease. Here we report the specific action of the peptide encompassing the 1-14 sequence of the human NGF (NGF(1-14)), identified on the basis of scattered indications present in literature. The biological activity of NGF(1-14) was tested on PC12 cells, and its binding with TrkA was predicted by means of a computational approach. NGF(1-14) does not elicit the neurite outgrowth promoting activity, typical of the whole protein, and it only has a moderate action on PC12 proliferation. However, this peptide exerts, in a dose and time dependent fashion, an effective and specific NGF-like action on some highly conserved and biologically crucial intermediates of its intracellular targets such as Akt and CREB. These findings indicate that not all TrkA pathways must be at all times operative, and open the possibility of testing each of them in relation with specific NGF needs, biological actions, and potential therapeutic use.
Collapse
Affiliation(s)
- Alessio Travaglia
- Center for Neural Science, New York University, 4 Washington Place, New York, New York 10003, United States
| | - Adriana Pietropaolo
- Dipartimento di Scienze della Salute, Università di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Rossana Di Martino
- Istituto di Bioimmagini e Fisiologia Molecolare (IBFM)-CNR, C.da Pietrapollastra-Pisciotto, Cefalù, Palermo 90015, Italy
| | - Vincenzo G. Nicoletti
- Dipartimento di Scienze Biomediche e Biotecnologiche - Sezione di Biochimica Medica, Università degli Studi di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
- Istituto Nazionale Biostrutture e Biosistemi (INBB) − Sezione Biomolecole, Consorzio Interuniversitario, Viale Medaglie d’Oro 305, 00136 Roma, Italy
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy
| | - Pietro Calissano
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano, 64-65, 00143 Rome, Italy
| | | |
Collapse
|
49
|
Marchetti L, Luin S, Bonsignore F, de Nadai T, Beltram F, Cattaneo A. Ligand-induced dynamics of neurotrophin receptors investigated by single-molecule imaging approaches. Int J Mol Sci 2015; 16:1949-79. [PMID: 25603178 PMCID: PMC4307343 DOI: 10.3390/ijms16011949] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 01/05/2015] [Indexed: 01/14/2023] Open
Abstract
Neurotrophins are secreted proteins that regulate neuronal development and survival, as well as maintenance and plasticity of the adult nervous system. The biological activity of neurotrophins stems from their binding to two membrane receptor types, the tropomyosin receptor kinase and the p75 neurotrophin receptors (NRs). The intracellular signalling cascades thereby activated have been extensively investigated. Nevertheless, a comprehensive description of the ligand-induced nanoscale details of NRs dynamics and interactions spanning from the initial lateral movements triggered at the plasma membrane to the internalization and transport processes is still missing. Recent advances in high spatio-temporal resolution imaging techniques have yielded new insight on the dynamics of NRs upon ligand binding. Here we discuss requirements, potential and practical implementation of these novel approaches for the study of neurotrophin trafficking and signalling, in the framework of current knowledge available also for other ligand-receptor systems. We shall especially highlight the correlation between the receptor dynamics activated by different neurotrophins and the respective signalling outcome, as recently revealed by single-molecule tracking of NRs in living neuronal cells.
Collapse
Affiliation(s)
- Laura Marchetti
- National Enterprise for nanoScience and nanoTechnology (NEST) Laboratory, Scuola Normale Superiore and Istituto Nanoscienze-CNR, Piazza San Silvestro 12, Pisa I-56127, Italy.
| | - Stefano Luin
- National Enterprise for nanoScience and nanoTechnology (NEST) Laboratory, Scuola Normale Superiore and Istituto Nanoscienze-CNR, Piazza San Silvestro 12, Pisa I-56127, Italy.
| | - Fulvio Bonsignore
- National Enterprise for nanoScience and nanoTechnology (NEST) Laboratory, Scuola Normale Superiore and Istituto Nanoscienze-CNR, Piazza San Silvestro 12, Pisa I-56127, Italy.
| | - Teresa de Nadai
- Biology Laboratory (BioSNS), Scuola Normale Superiore and Istituto di Neuroscienze-CNR, via Moruzzi 1, Pisa I-56100, Italy.
| | - Fabio Beltram
- National Enterprise for nanoScience and nanoTechnology (NEST) Laboratory, Scuola Normale Superiore and Istituto Nanoscienze-CNR, Piazza San Silvestro 12, Pisa I-56127, Italy.
| | - Antonino Cattaneo
- Biology Laboratory (BioSNS), Scuola Normale Superiore and Istituto di Neuroscienze-CNR, via Moruzzi 1, Pisa I-56100, Italy.
| |
Collapse
|
50
|
Mohamed AR, El-Hadidy WF. Effect of orexin-A (hypocretin-1) on hyperalgesic and cachectic manifestations of experimentally induced rheumatoid arthritis in rats. Can J Physiol Pharmacol 2014; 92:813-20. [DOI: 10.1139/cjpp-2014-0258] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Orexin-A has been shown to modulate pain sensation and increase appetite. Rheumatoid arthritis (RA) is characterized by joint destruction, deformity, hyperalgesia, and weight reduction. Aim: Evaluate the possible effect of orexin-A on hyperalgesic and cachectic manifestations in an adjuvant-induced arthritis (AIA) rat model. Methods: Forty adult male Wistar rats were distributed among 4 groups; I, normal controls; II, rats with AIA induced by intradermal injection of Mycobacterium butyricum, but with no other treatment; III, AIA rats treated daily with an intravenous injection of orexin-A for 8 days; and IV, AIA rats treated orally with dexamethasone for 8 days. The parameters we assessed were pain-associated behavior, body mass, hind paw volume, serum levels of nerve growth factor (NGF) and neuropeptide Y (NPY). Results: Orexin-A caused a significant reduction in pain sensation and NGF levels, and increased body mass and the levels of NPY, whereas treatment with dexamethasone led to significant reductions in paw swelling and pain sensation. Conclusion: Orexin-A has hypoalgesic properties and increases body mass, whereas dexamethasone has a potent anti-inflammatory effect. Therefore, the combination of orexin-A and dexamethasone should have a greater effect with respect to attenuating the manifestations and complications associated with RA.
Collapse
Affiliation(s)
- Adham R. Mohamed
- Physiology Department, Faculty of Medicine, Alexandria University, Egypt
| | - Wessam F. El-Hadidy
- Pharmacology and Experimental Therapeutics Department, Medical Research Institute, Alexandria University, 165 Horrya Avenue, Egypt
| |
Collapse
|