1
|
Xu Y, Liu Z, Xu J, Xu L, He Z, Liu F, Wang Y. Role of brain-derived neurotrophic factor in frailty: From mechanisms to interventions. Biomed Pharmacother 2025; 186:118016. [PMID: 40187046 DOI: 10.1016/j.biopha.2025.118016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025] Open
Abstract
Frailty is a common medical syndrome which largely increases the risk of disability, depression, falls, hospitalization and mortality. An increasing number of research suggests that frailty is reversible by medical interventions at its early stage. Therefore, efficient detection is utterly important for frail population. Since numerous biological processes have been indicated in frail population, the critical regulators in these biological processes could provide biomarkers for early detection or treatment for frailty. The brain-derived neurotrophic factor (BDNF) has been associated with several biological process ranging from cognitive function to inflammation, therefore it could be an important regulator for frailty. In this review, we would discuss the mechanism association between different indicators of frailty and BDNF. Furthermore, we summarize the approaches to interfere with BDNF in healthy and pathologic condition, which could lead to identification of potential interventional strategies for frailty.
Collapse
Affiliation(s)
- Yuanchun Xu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Ziyan Liu
- Department of Nursing, Traditional Chinese Medicine Hospital of Tongliang, Tongliang Chongqing 402560, China
| | - Jiao Xu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Lunshan Xu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Zongsheng He
- Department of Gastroenterology, Daping Hospital,Army Medical University, Chongqing 400042, China
| | - Fang Liu
- Department of Nursing, Traditional Chinese Medicine Hospital of Tongliang, Tongliang Chongqing 402560, China.
| | - Yaling Wang
- Department of Nursing, Daping Hospital, Army Medical University, Chongqing 400042, China.
| |
Collapse
|
2
|
Balanyà-Segura M, Polishchuk A, Just-Borràs L, Cilleros-Mañé V, Silvera C, Jami-ElHirchi M, Pinent M, Ardévol A, Tomàs M, Lanuza MA, Hurtado E, Tomàs J. Protective effects of grape seed procyanidin extract on neurotrophic and muscarinic signaling pathways in the aging neuromuscular junction. Food Funct 2025. [PMID: 40231589 DOI: 10.1039/d5fo00286a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
At the neuromuscular junction (NMJ), which coordinates movement, postsynaptic-derived neurotrophic factors have neuroprotective functions and retrogradely regulate the exocytotic machinery involved in neurotransmitter release. In parallel, presynaptic autocrine muscarinic signaling plays a fundamental modulatory role in this synapse. We previously found that these signaling pathways are impaired in the aged neuromuscular system. In this follow-up study, we investigated an anti-aging strategy using grape seed procyanidin extract (GSPE), a common dietary antioxidant known for its neuroprotective properties in various pathologies, but its effects on the aged neuromuscular system remain unexplored. This study analyses whether GSPE can mitigate age-associated impairments in neurotrophic and muscarinic signaling within the neuromuscular system. We assessed the expression (protein levels) and activation (phosphorylation) of the key proteins in the brain-derived-neurotrophic-factor (BDNF)/neurotrophin 4 (NT-4) and muscarinic pathways in the extensor digitorum longus (EDL) muscles of aged rats, with comparisons to GSPE-treated aged rats and young controls. The results demonstrate that GSPE treatment prevents the most relevant aging-induced changes in neurotrophic and muscarinic receptor isoforms, downstream protein kinases, and their targets in the neurotransmitter exocytotic machinery. Nevertheless, GSPE was less effective at preventing alterations in some other proteins within these pathways, such as calcium channels, and did not modify several other molecules involved in these pathways, which remain unchanged during aging. Overall, this study highlights the neuroprotective potential of GSPE in preventing fundamental age-related molecular changes at the NMJ, which helps improve functionality and may increase the quality of life and lifespan in aged individuals.
Collapse
Affiliation(s)
- Marta Balanyà-Segura
- Universitat Rovira i Virgili, Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Sant Llorenç 21, 43201 Reus, Spain.
| | - Aleksandra Polishchuk
- Universitat Rovira i Virgili, Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Sant Llorenç 21, 43201 Reus, Spain.
| | - Laia Just-Borràs
- Universitat Rovira i Virgili, Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Sant Llorenç 21, 43201 Reus, Spain.
| | - Víctor Cilleros-Mañé
- Universitat Rovira i Virgili, Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Sant Llorenç 21, 43201 Reus, Spain.
| | - Carolina Silvera
- Universitat Rovira i Virgili, Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Sant Llorenç 21, 43201 Reus, Spain.
| | - Meryem Jami-ElHirchi
- Universitat Rovira i Virgili, Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Sant Llorenç 21, 43201 Reus, Spain.
| | - Montserrat Pinent
- Universitat Rovira i Virgili, MoBioFood Research Group, Campus Sescelades, Marcel.lí Domingo 1, 43007 Tarragona, Spain.
| | - Anna Ardévol
- Universitat Rovira i Virgili, MoBioFood Research Group, Campus Sescelades, Marcel.lí Domingo 1, 43007 Tarragona, Spain.
| | - Marta Tomàs
- Universitat Rovira i Virgili, Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Sant Llorenç 21, 43201 Reus, Spain.
| | - Maria A Lanuza
- Universitat Rovira i Virgili, Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Sant Llorenç 21, 43201 Reus, Spain.
| | - Erica Hurtado
- Universitat Rovira i Virgili, Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Sant Llorenç 21, 43201 Reus, Spain.
| | - Josep Tomàs
- Universitat Rovira i Virgili, Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Sant Llorenç 21, 43201 Reus, Spain.
| |
Collapse
|
3
|
Tomàs J, Cilleros-Mañé V, Just-Borràs L, Balanyà-Segura M, Polishchuk A, Nadal L, Tomàs M, Silvera-Simón C, Santafé MM, Lanuza MA. Brain-derived neurotrophic factor signaling in the neuromuscular junction during developmental axonal competition and synapse elimination. Neural Regen Res 2025; 20:394-401. [PMID: 38819042 PMCID: PMC11317955 DOI: 10.4103/1673-5374.391314] [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: 07/25/2023] [Revised: 10/25/2023] [Accepted: 11/16/2023] [Indexed: 06/01/2024] Open
Abstract
During the development of the nervous system, there is an overproduction of neurons and synapses. Hebbian competition between neighboring nerve endings and synapses performing different activity levels leads to their elimination or strengthening. We have extensively studied the involvement of the brain-derived neurotrophic factor-Tropomyosin-related kinase B receptor neurotrophic retrograde pathway, at the neuromuscular junction, in the axonal development and synapse elimination process versus the synapse consolidation. The purpose of this review is to describe the neurotrophic influence on developmental synapse elimination, in relation to other molecular pathways that we and others have found to regulate this process. In particular, we summarize our published results based on transmitter release analysis and axonal counts to show the different involvement of the presynaptic acetylcholine muscarinic autoreceptors, coupled to downstream serine-threonine protein kinases A and C (PKA and PKC) and voltage-gated calcium channels, at different nerve endings in developmental competition. The dynamic changes that occur simultaneously in several nerve terminals and synapses converge across a postsynaptic site, influence each other, and require careful studies to individualize the mechanisms of specific endings. We describe an activity-dependent balance (related to the extent of transmitter release) between the presynaptic muscarinic subtypes and the neurotrophin-mediated TrkB/p75NTR pathways that can influence the timing and fate of the competitive interactions between the different axon terminals. The downstream displacement of the PKA/PKC activity ratio to lower values, both in competing nerve terminals and at postsynaptic sites, plays a relevant role in controlling the elimination of supernumerary synapses. Finally, calcium entry through L- and P/Q- subtypes of voltage-gated calcium channels (both channels are present, together with the N-type channel in developing nerve terminals) contributes to reduce transmitter release and promote withdrawal of the most unfavorable nerve terminals during elimination (the weakest in acetylcholine release and those that have already become silent). The main findings contribute to a better understanding of punishment-rewarding interactions between nerve endings during development. Identifying the molecular targets and signaling pathways that allow synapse consolidation or withdrawal of synapses in different situations is important for potential therapies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Josep Tomàs
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Víctor Cilleros-Mañé
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Laia Just-Borràs
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Marta Balanyà-Segura
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Aleksandra Polishchuk
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Laura Nadal
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Marta Tomàs
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Carolina Silvera-Simón
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Manel M. Santafé
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Maria A. Lanuza
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
4
|
Li Y, Badawi Y, Meriney SD. Age-Related Homeostatic Plasticity at Rodent Neuromuscular Junctions. Cells 2024; 13:1684. [PMID: 39451202 PMCID: PMC11506802 DOI: 10.3390/cells13201684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Motor ability decline remains a major threat to the quality of life of the elderly. Although the later stages of aging co-exist with degenerative pathologies, the long process of aging is more complicated than a simple and gradual degeneration. To combat senescence and the associated late-stage degeneration of the neuromuscular system, it is imperative to examine changes that occur during the long process of aging. Prior to late-stage degeneration, age-induced changes in the neuromuscular system trigger homeostatic plasticity. This unique phenomenon may be important for the maintenance of the neuromuscular system during the early stages of aging. In this review, we will focus on age-induced changes in neurotransmission at the neuromuscular junction, providing the potential mechanisms responsible for these changes. The goal is to highlight these key elements and their role in regulating neurotransmission, facilitating future research efforts to combat late-stage degeneration in the neuromuscular system by preserving the functional and structural integrity of these elements prior to the late stage of aging.
Collapse
Affiliation(s)
| | | | - Stephen D. Meriney
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA; (Y.L.); (Y.B.)
| |
Collapse
|
5
|
Zhu X, Chen W, Thirupathi A. Sprint Interval Training Improves Brain-Derived Neurotropic Factor-Induced Benefits in Brain Health-A Possible Molecular Signaling Intervention. BIOLOGY 2024; 13:562. [PMID: 39194500 DOI: 10.3390/biology13080562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Physical exercise can significantly impact our bodies, affecting our functional capacity, structure establishment, and molecular makeup. The magnitude of these changes depends on the specific exercise protocols used. For instance, low-to-moderate-intensity exercise can activate important molecular targets in the short term, such as BDNF-mediated signaling, while high-intensity exercise can maintain these signaling molecules in the active state for a longer term. This makes it challenging to recommend specific exercises for obtaining BDNF-induced benefits. Additionally, exercise-induced molecular signaling targets can have positive and negative effects, with some exercises blunting these targets and others activating them. For example, increasing BDNF concentration through exercise can be beneficial for brain health, but it may also have a negative impact on conditions such as bipolar disorder. Therefore, a deeper understanding of a specific exercise-mediated mechanistic approach is required. This review will delve into how the sprint exercise-mediated activation of BDNF could help maintain brain health and explore potential molecular interventions.
Collapse
Affiliation(s)
- Xueqiang Zhu
- School of Competitive Sports, Shandong Sport University, Rizhao 276826, China
| | - Wenjia Chen
- School of Physical Education, China University of Mining and Technology, Xuzhou 221116, China
| | - Anand Thirupathi
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China
| |
Collapse
|
6
|
Polishchuk A, Cilleros-Mañé V, Balanyà-Segura M, Just-Borràs L, Forniés-Mariné A, Silvera-Simón C, Tomàs M, Jami El Hirchi M, Hurtado E, Tomàs J, Lanuza MA. BDNF/TrkB signalling, in cooperation with muscarinic signalling, retrogradely regulates PKA pathway to phosphorylate SNAP-25 and Synapsin-1 at the neuromuscular junction. Cell Commun Signal 2024; 22:371. [PMID: 39044222 PMCID: PMC11265447 DOI: 10.1186/s12964-024-01735-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/04/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Protein kinase A (PKA) enhances neurotransmission at the neuromuscular junction (NMJ), which is retrogradely regulated by nerve-induced muscle contraction to promote Acetylcholine (ACh) release through the phosphorylation of molecules involved in synaptic vesicle exocytosis (SNAP-25 and Synapsin-1). However, the molecular mechanism of the retrograde regulation of PKA subunits and its targets by BDNF/TrkB pathway and muscarinic signalling has not been demonstrated until now. At the NMJ, retrograde control is mainly associated with BDNF/TrkB signalling as muscle contraction enhances BDNF levels and controls specific kinases involved in the neurotransmission. Neurotransmission at the NMJ is also highly modulated by muscarinic receptors M1 and M2 (mAChRs), which are related to PKA and TrkB signallings. Here, we investigated the hypothesis that TrkB, in cooperation with mAChRs, regulates the activity-dependent dynamics of PKA subunits to phosphorylate SNAP-25 and Synapsin-1. METHODS To explore this, we stimulated the rat phrenic nerve at 1Hz (30 minutes), with or without subsequent contraction (abolished by µ-conotoxin GIIIB). Pharmacological treatments were conducted with the anti-TrkB antibody clone 47/TrkB for TrkB inhibition and exogenous h-BDNF; muscarinic inhibition with Pirenzepine-dihydrochloride and Methoctramine-tetrahydrochloride for M1 and M2 mAChRs, respectively. Diaphragm protein levels and phosphorylation' changes were detected by Western blotting. Location of the target proteins was demonstrated using immunohistochemistry. RESULTS While TrkB does not directly impact the levels of PKA catalytic subunits Cα and Cβ, it regulates PKA regulatory subunits RIα and RIIβ, facilitating the phosphorylation of critical exocytotic targets such as SNAP-25 and Synapsin-1. Furthermore, the muscarinic receptors pathway maintains a delicate balance in this regulatory process. These findings explain the dynamic interplay of PKA subunits influenced by BDNF/TrkB signalling, M1 and M2 mAChRs pathways, that are differently regulated by pre- and postsynaptic activity, demonstrating the specific roles of the BDNF/TrkB and muscarinic receptors pathway in retrograde regulation. CONCLUSION This complex molecular interplay has the relevance of interrelating two fundamental pathways in PKA-synaptic modulation: one retrograde (neurotrophic) and the other autocrine (muscarinic). This deepens the fundamental understanding of neuromuscular physiology of neurotransmission that gives plasticity to synapses and holds the potential for identifying therapeutic strategies in conditions characterized by impaired neuromuscular communication.
Collapse
Affiliation(s)
- Aleksandra Polishchuk
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Víctor Cilleros-Mañé
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Marta Balanyà-Segura
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Laia Just-Borràs
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Anton Forniés-Mariné
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
| | - Carolina Silvera-Simón
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Marta Tomàs
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Meryem Jami El Hirchi
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Erica Hurtado
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Josep Tomàs
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Maria A Lanuza
- Universitat Rovira i Virgili. Unitat d'Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut. c/ Sant Llorenç 21, Reus, 43201, Spain.
- Unitat d'Histologia i Neurobiologia (UHNeurob), Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain.
| |
Collapse
|
7
|
Balanyà-Segura M, Polishchuk A, Just-Borràs L, Cilleros-Mañé V, Silvera C, Ardévol A, Tomàs M, Lanuza MA, Hurtado E, Tomàs J. Molecular Adaptations of BDNF/NT-4 Neurotrophic and Muscarinic Pathways in Ageing Neuromuscular Synapses. Int J Mol Sci 2024; 25:8018. [PMID: 39125587 PMCID: PMC11311581 DOI: 10.3390/ijms25158018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 08/12/2024] Open
Abstract
Age-related conditions, such as sarcopenia, cause physical disabilities for an increasing section of society. At the neuromuscular junction, the postsynaptic-derived neurotrophic factors brain-derived neurotrophic factor (BDNF) and neurotrophin 4 (NT-4) have neuroprotective functions and contribute to the correct regulation of the exocytotic machinery. Similarly, presynaptic muscarinic signalling plays a fundamental modulatory function in this synapse. However, whether or not these signalling pathways are compromised in ageing neuromuscular system has not yet been analysed. The present study analyses, through Western blotting, the differences in expression and activation of the main key proteins of the BDNF/NT-4 and muscarinic pathways related to neurotransmission in young versus ageing Extensor digitorum longus (EDL) rat muscles. The main results show an imbalance in several sections of these pathways: (i) a change in the stoichiometry of BDNF/NT-4, (ii) an imbalance of Tropomyosin-related kinase B receptor (TrkB)-FL/TrkB-T1 and neurotrophic receptor p 75 (p75NTR), (iii) no changes in the cytosol/membrane distribution of phosphorylated downstream protein kinase C (PKC)βI and PKCε, (iv) a reduction in the M2-subtype muscarinic receptor and P/Q-subtype voltage-gated calcium channel, (v) an imbalance of phosphorylated mammalian uncoordinated-18-1 (Munc18-1) (S313) and synaptosomal-associated protein 25 (SNAP-25) (S187), and (vi) normal levels of molecules related to the management of acetylcholine (Ach). Based on this descriptive analysis, we hypothesise that these pathways can be adjusted to ensure neurotransmission rather than undergoing negative alterations caused by ageing. However, further studies are needed to assess this hypothetical suggestion. Our results contribute to the understanding of some previously described neuromuscular functional age-related impairments. Strategies to promote these signalling pathways could improve the neuromuscular physiology and quality of life of older people.
Collapse
Affiliation(s)
- Marta Balanyà-Segura
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.B.-S.); (A.P.); (L.J.-B.); (V.C.-M.); (C.S.); (M.T.); (J.T.)
| | - Aleksandra Polishchuk
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.B.-S.); (A.P.); (L.J.-B.); (V.C.-M.); (C.S.); (M.T.); (J.T.)
| | - Laia Just-Borràs
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.B.-S.); (A.P.); (L.J.-B.); (V.C.-M.); (C.S.); (M.T.); (J.T.)
| | - Víctor Cilleros-Mañé
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.B.-S.); (A.P.); (L.J.-B.); (V.C.-M.); (C.S.); (M.T.); (J.T.)
| | - Carolina Silvera
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.B.-S.); (A.P.); (L.J.-B.); (V.C.-M.); (C.S.); (M.T.); (J.T.)
| | - Anna Ardévol
- MoBioFood Research Group, Campus Sescelades, Universitat Rovira i Virgili, Marcel.lí Domingo 1, 43007 Tarragona, Spain;
| | - Marta Tomàs
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.B.-S.); (A.P.); (L.J.-B.); (V.C.-M.); (C.S.); (M.T.); (J.T.)
| | - Maria A. Lanuza
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.B.-S.); (A.P.); (L.J.-B.); (V.C.-M.); (C.S.); (M.T.); (J.T.)
| | - Erica Hurtado
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.B.-S.); (A.P.); (L.J.-B.); (V.C.-M.); (C.S.); (M.T.); (J.T.)
| | - Josep Tomàs
- Unitat d’Histologia i Neurobiologia (UHNeurob), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201 Reus, Spain; (M.B.-S.); (A.P.); (L.J.-B.); (V.C.-M.); (C.S.); (M.T.); (J.T.)
| |
Collapse
|
8
|
Deng C, Chen H. Brain-derived neurotrophic factor/tropomyosin receptor kinase B signaling in spinal muscular atrophy and amyotrophic lateral sclerosis. Neurobiol Dis 2024; 190:106377. [PMID: 38092270 DOI: 10.1016/j.nbd.2023.106377] [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: 09/17/2023] [Revised: 11/15/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023] Open
Abstract
Tropomyosin receptor kinase B (TrkB) and its primary ligand brain-derived neurotrophic factor (BDNF) are expressed in the neuromuscular system, where they affect neuronal survival, differentiation, and functions. Changes in BDNF levels and full-length TrkB (TrkB-FL) signaling have been revealed in spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS), two common forms of motor neuron diseases that are characterized by defective neuromuscular junctions in early disease stages and subsequently progressive muscle weakness. This review summarizes the current understanding of BDNF/TrkB-FL-related research in SMA and ALS, with an emphasis on their alterations in the neuromuscular system and possible BDNF/TrkB-FL-targeting therapeutic strategies. The limitations of current studies and future directions are also discussed, giving the hope of discovering novel and effective treatments.
Collapse
Affiliation(s)
- Chunchu Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
9
|
Tarasova E, Bogacheva P, Chernyshev K, Balezina O. Quantal size increase induced by the endocannabinoid 2-arachidonoylglycerol requires activation of CGRP receptors in mouse motor synapses. Synapse 2024; 78:e22281. [PMID: 37694983 DOI: 10.1002/syn.22281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/12/2023]
Abstract
In mouse motor synapses, the exogenous application of the endocannabinoid (EC) 2-arachidonoylglycerol (2-AG) increases acetylcholine (ACh) quantal size due to the activation of CB1 receptors and the stimulation of ACh vesicular uptake. In the present study, microelectrode recordings of miniature endplate potentials (MEPP) revealed that this effect of 2-AG is independent of brain-derived neurotrophic factor (BDNF) signaling but involves the activation of calcitonin gene-related peptide (CGRP) receptors along with CB1 receptors. Potentiation of MEPP amplitude in the presence of 2-AG was prevented by blockers of CGRP receptors and ryanodine receptors (RyR) and by inhibitors of phospholipase C (PLC) and Ca2+ /calmodulin-dependent protein kinase II (CaMKII). Therefore, we suggest a hypothetical chain of events, which starts from the activation of presynaptic CB1 receptors, involves PLC, RyR, and CaMKII, and results in CGRP release with the subsequent activation of presynaptic CGRP receptors. Activation of CGRP receptors is probably a part of a complex molecular cascade leading to the 2-AG-induced increase in ACh quantal size and MEPP amplitude. We propose that the same chain of events may also take place if 2-AG is endogenously produced in mouse motor synapses, because the increase in MEPP amplitude that follows after prolonged tetanic muscle contractions (30 Hz, 2 min) was prevented by the blocking of CB1 receptors. This work may help to unveil the previously unknown aspects of the functional interaction between ECs and peptide modulators aimed at the regulation of quantal size and synaptic transmission.
Collapse
Affiliation(s)
- Ekaterina Tarasova
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Polina Bogacheva
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Kirill Chernyshev
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Olga Balezina
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russian Federation
| |
Collapse
|
10
|
Fogarty MJ, Dasgupta D, Khurram OU, Sieck GC. Chemogenetic inhibition of TrkB signalling reduces phrenic motor neuron survival and size. Mol Cell Neurosci 2023; 125:103847. [PMID: 36958643 PMCID: PMC10247511 DOI: 10.1016/j.mcn.2023.103847] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 03/07/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023] Open
Abstract
Brain derived neurotrophic factor (BDNF) signalling through its high-affinity tropomyosin receptor kinase B (TrkB) is known to have potent effects on motor neuron survival and morphology during development and in neurodegenerative diseases. Here, we employed a novel 1NMPP1 sensitive TrkBF616 rat model to evaluate the effect of 14 days inhibition of TrkB signalling on phrenic motor neurons (PhMNs). Adult female and male TrkBF616 rats were divided into 1NMPP1 or vehicle treated groups. Three days prior to treatment, PhMNs in both groups were initially labeled via intrapleural injection of Alexa-Fluor-647 cholera toxin B (CTB). After 11 days of treatment, retrograde axonal uptake/transport was assessed by secondary labeling of PhMNs by intrapleural injection of Alexa-Fluor-488 CTB. After 14 days of treatment, the spinal cord was excised 100 μm thick spinal sections containing PhMNs were imaged using two-channel confocal microscopy. TrkB inhibition reduced the total number of PhMNs by ∼16 %, reduced the mean PhMN somal surface areas by ∼25 %, impaired CTB uptake 2.5-fold and reduced the estimated PhMN dendritic surface area by ∼38 %. We conclude that inhibition of TrkB signalling alone in adult TrkBF616 rats is sufficient to lead to PhMN loss, morphological degeneration and deficits in retrograde axonal uptake/transport.
Collapse
Affiliation(s)
- Matthew J Fogarty
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Debanjali Dasgupta
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Obaid U Khurram
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Gary C Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
11
|
Gerwin RD. A New Unified Theory of Trigger Point Formation: Failure of Pre- and Post-Synaptic Feedback Control Mechanisms. Int J Mol Sci 2023; 24:ijms24098142. [PMID: 37175845 PMCID: PMC10179372 DOI: 10.3390/ijms24098142] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
The origin of the myofascial trigger point (TrP), an anomalous locus in muscle, has never been well-described. A new trigger point hypothesis (the new hypothesis) presented here addresses this lack. The new hypothesis is based on the concept that existing myoprotective feedback mechanisms that respond to muscle overactivity, low levels of adenosine triphosphate, (ATP) or a low pH, fail to protect muscle in certain circumstances, such as intense muscle activity, resulting in an abnormal accumulation of intracellular Ca2+, persistent actin-myosin cross bridging, and then activation of the nociceptive system, resulting in the formation of a trigger point. The relevant protective feedback mechanisms include pre- and postsynaptic sympathetic nervous system modulation, modulators of acetylcholine release at the neuromuscular junction, and mutations/variants or post-translational functional alterations in either of two ion channelopathies, the ryanodine receptor and the potassium-ATP ion channel, both of which exist in multiple mutation states that up- or downregulate ion channel function. The concepts that are central to the origin of at least some TrPs are the failure of protective feedback mechanisms and/or of certain ion channelopathies that are new concepts in relation to myofascial trigger points.
Collapse
Affiliation(s)
- Robert D Gerwin
- Department of Neurology, The Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
12
|
Ishida J, Kato A. Recent Advances in the Nutritional Screening, Assessment, and Treatment of Japanese Patients on Hemodialysis. J Clin Med 2023; 12:jcm12062113. [PMID: 36983116 PMCID: PMC10051275 DOI: 10.3390/jcm12062113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/19/2023] [Accepted: 03/04/2023] [Indexed: 03/30/2023] Open
Abstract
Patients on hemodialysis (HD) have a higher rate of protein-energy wasting (PEW) due to lower dietary intake of energy and protein (particularly on dialysis days) and greater loss of many nutrients in the dialysate effluent than other patients. The most well-known method of nutritional screening is the subjective global assessment. Moreover, the Global Leadership Initiative on MalnutIrition has developed the first internationally standardized method for diagnosing malnutrition; however, its use in patients on HD has not been established. In contrast, the nutritional risk index for Japanese patients on HD has recently been developed as a screening tool for malnutrition in patients on HD, based on the modified PEW criteria. These tools are beneficial for screening nutritional disorders, enabling registered dietitians to assess patients' dietary intake on dialysis and non-dialysis days and provide advice on dietary intake, especially immediately after dialysis cessation. Oral supplementation with enteral nutrients containing whey protein may also be administered when needed. In patients that experience adverse effects from oral supplementation, intradialytic parenteral nutrition (IDPN) should be combined with moderate dietary intake because IDPN alone cannot provide sufficient nutrition.
Collapse
Affiliation(s)
- Junko Ishida
- Department of Food and Nutritional Environment, College of Human Life and Environment, Kinjo Gakuin University, Nagoya 463-8521, Japan
| | - Akihiko Kato
- Blood Purification Unit, Hamamatsu University Hospital, Hamamatsu 431-3192, Japan
| |
Collapse
|
13
|
Polishchuk A, Cilleros-Mañé V, Just-Borràs L, Balanyà-Segura M, Vandellòs Pont G, Silvera Simón C, Tomàs M, Garcia N, Tomàs J, Lanuza MA. Synaptic retrograde regulation of the PKA-induced SNAP-25 and Synapsin-1 phosphorylation. Cell Mol Biol Lett 2023; 28:17. [PMID: 36869288 PMCID: PMC9985302 DOI: 10.1186/s11658-023-00431-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/09/2023] [Indexed: 03/05/2023] Open
Abstract
BACKGROUND Bidirectional communication between presynaptic and postsynaptic components contribute to the homeostasis of the synapse. In the neuromuscular synapse, the arrival of the nerve impulse at the presynaptic terminal triggers the molecular mechanisms associated with ACh release, which can be retrogradely regulated by the resulting muscle contraction. This retrograde regulation, however, has been poorly studied. At the neuromuscular junction (NMJ), protein kinase A (PKA) enhances neurotransmitter release, and the phosphorylation of the molecules of the release machinery including synaptosomal associated protein of 25 kDa (SNAP-25) and Synapsin-1 could be involved. METHODS Accordingly, to study the effect of synaptic retrograde regulation of the PKA subunits and its activity, we stimulated the rat phrenic nerve (1 Hz, 30 min) resulting or not in contraction (abolished by µ-conotoxin GIIIB). Changes in protein levels and phosphorylation were detected by western blotting and cytosol/membrane translocation by subcellular fractionation. Synapsin-1 was localized in the levator auris longus (LAL) muscle by immunohistochemistry. RESULTS Here we show that synaptic PKA Cβ subunit regulated by RIIβ or RIIα subunits controls activity-dependent phosphorylation of SNAP-25 and Synapsin-1, respectively. Muscle contraction retrogradely downregulates presynaptic activity-induced pSynapsin-1 S9 while that enhances pSNAP-25 T138. Both actions could coordinately contribute to decreasing the neurotransmitter release at the NMJ. CONCLUSION This provides a molecular mechanism of the bidirectional communication between nerve terminals and muscle cells to balance the accurate process of ACh release, which could be important to characterize molecules as a therapy for neuromuscular diseases in which neuromuscular crosstalk is impaired.
Collapse
Affiliation(s)
- Aleksandra Polishchuk
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Víctor Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Laia Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Marta Balanyà-Segura
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Genís Vandellòs Pont
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Carolina Silvera Simón
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain
| | - Josep Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain.
| | - Maria A Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201, Reus, Spain.
| |
Collapse
|
14
|
Just-Borràs L, Cilleros-Mañé V, Polishchuk A, Balanyà-Segura M, Tomàs M, Garcia N, Tomàs J, Lanuza MA. TrkB signaling is correlated with muscular fatigue resistance and less vulnerability to neurodegeneration. Front Mol Neurosci 2022; 15:1069940. [PMID: 36618825 PMCID: PMC9813967 DOI: 10.3389/fnmol.2022.1069940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
At the neuromuscular junction (NMJ), motor neurons and myocytes maintain a bidirectional communication that guarantees adequate functionality. Thus, motor neurons' firing pattern, which is influenced by retrograde muscle-derived neurotrophic factors, modulates myocyte contractibility. Myocytes can be fast-twitch fibers and become easily fatigued or slow-twitch fibers and resistant to fatigue. Extraocular muscles (EOM) show mixed properties that guarantee fast contraction speed and resistance to fatigue and the degeneration caused by Amyotrophic lateral sclerosis (ALS) disease. The TrkB signaling is an activity-dependent pathway implicated in the NMJ well-functioning. Therefore, it could mediate the differences between fast and slow myocytes' resistance to fatigue. The present study elucidates a specific protein expression profile concerning the TrkB signaling that correlates with higher resistance to fatigue and better neuroprotective capacity through time. The results unveil that Extra-ocular muscles (EOM) express lower levels of NT-4 that extend TrkB signaling, differential PKC expression, and a higher abundance of phosphorylated synaptic proteins that correlate with continuous neurotransmission requirements. Furthermore, common molecular features between EOM and slow soleus muscles including higher neurotrophic consumption and classic and novel PKC isoforms balance correlate with better preservation of these two muscles in ALS. Altogether, higher resistance of Soleus and EOM to fatigue and ALS seems to be associated with specific protein levels concerning the TrkB neurotrophic signaling.
Collapse
|
15
|
BDNF Spinal Overexpression after Spinal Cord Injury Partially Protects Soleus Neuromuscular Junction from Disintegration, Increasing VAChT and AChE Transcripts in Soleus but Not Tibialis Anterior Motoneurons. Biomedicines 2022; 10:biomedicines10112851. [DOI: 10.3390/biomedicines10112851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/20/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
After spinal cord transection (SCT) the interaction between motoneurons (MNs) and muscle is impaired, due to reorganization of the spinal network after a loss of supraspinal inputs. Rats subjected to SCT, treated with intraspinal injection of a AAV-BDNF (brain-derived neurotrophic factor) construct, partially regained the ability to walk. The central effects of this treatment have been identified, but its impact at the neuromuscular junction (NMJ) has not been characterized. Here, we compared the ability of NMJ pre- and postsynaptic machinery in the ankle extensor (Sol) and flexor (TA) muscles to respond to intraspinal AAV-BDNF after SCT. The gene expression of cholinergic molecules (VAChT, ChAT, AChE, nAChR, mAChR) was investigated in tracer-identified, microdissected MN perikarya, and in muscle fibers with the use of qPCR. In the NMJs, a distribution of VAChT, nAChR and Schwann cells was studied by immunofluorescence, and of synaptic vesicles and membrane active zones by electron microscopy. We showed partial protection of the Sol NMJs from disintegration, and upregulation of the VAChT and AChE transcripts in the Sol, but not the TA MNs after spinal enrichment with BDNF. We propose that the observed discrepancy in response to BDNF treatment is an effect of difference in the TrkB expression setting BDNF responsiveness, and of BDNF demands in Sol and TA muscles.
Collapse
|
16
|
Li Y, Li F, Qin D, Chen H, Wang J, Wang J, Song S, Wang C, Wang Y, Liu S, Gao D, Wang ZH. The role of brain derived neurotrophic factor in central nervous system. Front Aging Neurosci 2022; 14:986443. [PMID: 36158555 PMCID: PMC9493475 DOI: 10.3389/fnagi.2022.986443] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/23/2022] [Indexed: 11/15/2022] Open
Abstract
Brain derived neurotrophic factor (BDNF) has multiple biological functions which are mediated by the activation of two receptors, tropomyosin receptor kinase B (TrkB) receptor and the p75 neurotrophin receptor, involving in physiological and pathological processes throughout life. The diverse presence and activity of BDNF indicate its potential role in the pathogenesis, progression and treatment of both neurological and psychiatric disorders. This review is to provide a comprehensive assessment of the current knowledge and future directions in BDNF-associated research in the central nervous system (CNS), with an emphasis on the physiological and pathological functions of BDNF as well as its potential treatment effects in CNS diseases, including depression, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, and cerebral ischemic stroke.
Collapse
Affiliation(s)
- Yiyi Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fang Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dongdong Qin
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongyu Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jianhao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiabei Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shafei Song
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yamei Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Songyan Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dandan Gao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhi-Hao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Tosolini AP, Sleigh JN, Surana S, Rhymes ER, Cahalan SD, Schiavo G. BDNF-dependent modulation of axonal transport is selectively impaired in ALS. Acta Neuropathol Commun 2022; 10:121. [PMID: 35996201 PMCID: PMC9396851 DOI: 10.1186/s40478-022-01418-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 02/08/2023] Open
Abstract
Axonal transport ensures long-range delivery of essential cargoes between proximal and distal compartments, and is needed for neuronal development, function, and survival. Deficits in axonal transport have been detected at pre-symptomatic stages in the SOD1G93A and TDP-43M337V mouse models of amyotrophic lateral sclerosis (ALS), suggesting that impairments in this critical process are fundamental for disease pathogenesis. Strikingly, in ALS, fast motor neurons (FMNs) degenerate first whereas slow motor neurons (SMNs) are more resistant, and this is a currently unexplained phenomenon. The main aim of this investigation was to determine the effects of brain-derived neurotrophic factor (BDNF) on in vivo axonal transport in different α-motor neuron (MN) subtypes in wild-type (WT) and SOD1G93A mice. We report that despite displaying similar basal transport speeds, stimulation of wild-type MNs with BDNF enhances in vivo trafficking of signalling endosomes specifically in FMNs. This BDNF-mediated enhancement of transport was also observed in primary ventral horn neuronal cultures. However, FMNs display selective impairment of axonal transport in vivo in symptomatic SOD1G93A mice, and are refractory to BDNF stimulation, a phenotype that was also observed in primary embryonic SOD1G93A neurons. Furthermore, symptomatic SOD1G93A mice display upregulation of the classical non-pro-survival truncated TrkB and p75NTR receptors in muscles, sciatic nerves, and Schwann cells. Altogether, these data indicate that cell- and non-cell autonomous BDNF signalling is impaired in SOD1G93A MNs, thus identifying a new key deficit in ALS.
Collapse
Affiliation(s)
- Andrew P Tosolini
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK.
| | - James N Sleigh
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - Sunaina Surana
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - Elena R Rhymes
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK
| | - Stephen D Cahalan
- Comparative Neuromuscular Disease Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, University of London, London, NW1 0TU, UK
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK.
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
18
|
Rentería I, García-Suárez PC, Fry AC, Moncada-Jiménez J, Machado-Parra JP, Antunes BM, Jiménez-Maldonado A. The Molecular Effects of BDNF Synthesis on Skeletal Muscle: A Mini-Review. Front Physiol 2022; 13:934714. [PMID: 35874524 PMCID: PMC9306488 DOI: 10.3389/fphys.2022.934714] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
The brain-derived neurotrophic factor (BDNF) is a member of the nerve growth factor family which is generated mainly by the brain. Its main role involve synaptic modulation, neurogenesis, neuron survival, immune regulation, myocardial contraction, and angiogenesis in the brain. Together with the encephalon, some peripheral tissues synthesize BDNF like skeletal muscle. On this tissue, this neurotrophin participates on cellular mechanisms related to muscle function maintenance and plasticity as reported on recent scientific works. Moreover, during exercise stimuli the BDNF contributes directly to strengthening neuromuscular junctions, muscle regeneration, insulin-regulated glucose uptake and β-oxidation processes in muscle tissue. Given its vital relevance on many physiological mechanisms, the current mini-review focuses on discussing up-to-date knowledge about BDNF production in skeletal muscle and how this neurotrophin impacts skeletal muscle biology.
Collapse
Affiliation(s)
- I Rentería
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico
| | - P C García-Suárez
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico.,Department of Health, Sports and Exercise Sciences, University of Kansas, Lawrence, KS, United States
| | - A C Fry
- Department of Health, Sports and Exercise Sciences, University of Kansas, Lawrence, KS, United States
| | - J Moncada-Jiménez
- Human Movement Sciences Research Center (CIMOHU), University of Costa Rica, San José, Costa Rica
| | - J P Machado-Parra
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico
| | - B M Antunes
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico
| | - A Jiménez-Maldonado
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico
| |
Collapse
|
19
|
Azman KF, Zakaria R. Recent Advances on the Role of Brain-Derived Neurotrophic Factor (BDNF) in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:6827. [PMID: 35743271 PMCID: PMC9224343 DOI: 10.3390/ijms23126827] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 02/04/2023] Open
Abstract
Neurotrophins, such as brain-derived neurotrophic factor (BDNF), are essential for neuronal survival and growth. The signaling cascades initiated by BDNF and its receptor are the key regulators of synaptic plasticity, which plays important role in learning and memory formation. Changes in BDNF levels and signaling pathways have been identified in several neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Huntington's disease, and have been linked with the symptoms and course of these diseases. This review summarizes the current understanding of the role of BDNF in several neurodegenerative diseases, as well as the underlying molecular mechanism. The therapeutic potential of BDNF treatment is also discussed, in the hope of discovering new avenues for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Khairunnuur Fairuz Azman
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia;
| | | |
Collapse
|
20
|
Maroofi A, Bagheri Rouch A, Naderi N, Damirchi A. Effects of two different exercise paradigms on cardiac function, BDNF-TrkB expression, and myocardial protection in the presence and absence of Western diet. IJC HEART & VASCULATURE 2022; 40:101022. [PMID: 35399608 PMCID: PMC8991101 DOI: 10.1016/j.ijcha.2022.101022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/17/2022] [Accepted: 03/30/2022] [Indexed: 12/11/2022]
Abstract
Background Brain-derived neurotrophic factor (BDNF) -tropomyosin-related kinase receptor B (TrkB) signaling is a vital regulator of myocardial performance. Here, we tested the impact of high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) on heart function, metabolic parameters, and serum/cardiac BDNF (with its TrkB receptor) in animals fed a Western (WD) or regular diet (ND). Further, myocardial expression of pro-inflammatory cytokine interleukin-18 (IL-18) and cardioprotective molecule heme oxygens-1 (HO-1) were monitored. Methods Wistar rats were divided into HIIT, MICT, and sedentary (SED), all fed a WD or ND, for 12 weeks. Heart function, protein expression, and serum factors were assessed via echocardiography, western blotting, and ELISA, respectively. Results WD plus SED caused insulin resistance, dyslipidemia, visceral fat deposition, serum BDNF depletion as well as cardiac upregulation of IL-18 and downregulation of HO-1, without affecting, heart function and BDNF-TrkB expression. The cardiometabolic risk factors, serum BDNF losses, and IL-18 overexpression were similarly obviated by HIIT and MICT, although HO-1 expression was boosted by HIIT exclusively (even in ND). HIIT enhanced heart function, regardless of the diet. HIIT augmented cardiac BDNF expression, with a significant difference between ND and WD. Likewise, HIIT instigated TrkB expression only in ND. Conclusions HIIT and MICT can cope with myocardial inflammation and cardiometabolic risk factors in WD consumers and, exclusively, HIIT may grant further protection by increasing heart function, BDNF-TrkB expression, and HO-1 expression. Thus, the HIIT paradigm should be considered as a preference for subjects who require heart function to be preserved or enhanced.
Collapse
Affiliation(s)
- Abdulbaset Maroofi
- Department of Exercise Physiology, Faculty of Physical Education & Sport Sciences, University of Guilan, Rasht, Iran
| | - Ahmadreza Bagheri Rouch
- Department of Exercise Physiology, Faculty of Physical Education & Sport Sciences, University of Guilan, Rasht, Iran
| | - Nasim Naderi
- Rajaie Cardiovascular, Medical & Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Arsalan Damirchi
- Department of Exercise Physiology, Faculty of Physical Education & Sport Sciences, University of Guilan, Rasht, Iran
| |
Collapse
|
21
|
Arenas YM, Balzano T, Ivaylova G, Llansola M, Felipo V. The S1PR2-CCL2-BDNF-TrkB pathway mediates neuroinflammation and motor incoordination in hyperammonaemia. Neuropathol Appl Neurobiol 2022; 48:e12799. [PMID: 35152448 DOI: 10.1111/nan.12799] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 12/21/2021] [Accepted: 02/05/2022] [Indexed: 11/18/2024]
Abstract
AIMS Chronic hyperammonaemia and inflammation synergistically induce neurological impairment, including motor incoordination, in hepatic encephalopathy. Hyperammonaemic rats show neuroinflammation in the cerebellum which enhances GABAergic neurotransmission leading to motor incoordination. We aimed to identify underlying mechanisms. The aims were (1) to assess if S1PR2 is involved in microglial and astrocytic activation in the cerebellum of hyperammonaemic rats; (2) to identify pathways by which enhanced S1PR2 activation induces neuroinflammation and alters neurotransmission; (3) to assess if blocking S1PR2 reduces neuroinflammation and restores motor coordination in hyperammonaemic rats. METHODS We performed ex vivo studies in cerebellar slices from control or hyperammonaemic rats to identify pathways by which neuroinflammation enhances GABAergic neurotransmission in hyperammonaemia. Neuroinflammation and neurotransmission were assessed by immunochemistry/immunofluorescence and western blot. S1PR2 was blocked by intracerebral treatment with JTE-013 using osmotic mini-pumps. Motor coordination was assessed by beam walking. RESULTS Chronic hyperammonaemia enhances S1PR2 activation in the cerebellum by increasing its membrane expression. This increases CCL2, especially in Purkinje neurons. CCL2 activates CCR2 in microglia, leading to microglial activation, increased P2X4 membrane expression and BDNF in microglia. BDNF enhances TrkB activation in neurons, increasing KCC2 membrane expression. This enhances GABAergic neurotransmission, leading to motor incoordination in hyperammonaemic rats. Blocking S1PR2 in hyperammonaemic rats by intracerebral administration of JTE-013 normalises the S1PR2-CCL2-CCR2-BDNF-TrkB-KCC2 pathway, reduces glial activation and restores motor coordination in hyperammonaemic rats. CONCLUSIONS Enhanced S1PR2-CCL2-BDNF-TrkB pathway activation mediates neuroinflammation and incoordination in hyperammonaemia. The data raise a promising therapy for patients with hepatic encephalopathy using compounds targeting this pathway.
Collapse
Affiliation(s)
- Yaiza M Arenas
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Valencia, Spain
| | - Tiziano Balzano
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Valencia, Spain
| | - Gergana Ivaylova
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Valencia, Spain
| | - Marta Llansola
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Valencia, Spain
| |
Collapse
|
22
|
Effect of combined locomotor training and aerobic exercise on increasing handgrip strength in elderly with locomotive syndrome: A randomised controlled trial. Ann Med Surg (Lond) 2022; 78:103800. [PMID: 35734655 PMCID: PMC9207023 DOI: 10.1016/j.amsu.2022.103800] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 01/08/2023] Open
Abstract
Background Elderly with the locomotive syndrome is at high risk for fall and fractures. Thus multimodal therapy is needed to minimize the risk. Objective Analyzing the effect of combined locomotor training and aerobic exercise on muscle strength in elderly with locomotive syndrome stage 1. Methods This study used a pre-test and post-test design with 20 participants (treatment group = 10 participants and control group = 10 participants). The treatment group was given combined locomotor training and aerobic exercise, while the control group was only given aerobic exercise for eight weeks. Locomotor training was provided three times/week with progressive increase of set and repetition at each activity. Meanwhile, aerobic exercise was given seven times/week for 30 min per session. Participants were examined for muscle strength (handgrip strength) before and after the intervention. The analysis included paired t-test and an independent t-test with a p-value <0.05. Results The participants' mean age was 73.85 ± 4.75 years, with treatment group = 75.4 ± 4.88 years and control group = 72.3 ± 4.30 years (t = 1.508; 95% CI = −1.220 – 7420; p = 0.149). The HGS values in the treatment group were 13.89 ± 5.27 (pre-test) and 19.06 ± 4.54 (post-test; t = 11.765; 95% CI = −6.164 to −4.176; p < 0.001). Meanwhile, the HGS values in the control group at pre-test and post-test were 11.27 ± 2.17 and 13.03 ± 2.54, respectively (t = 2.057; 95% CI = −1.600 – 0.076; p = 0.070). The ΔHGS values of treatment and control group were 5.17 ± 1.39 and 1.76 ± 2.07, respectively (t = 4.329; 95% CI = 1.755–5.065; p < 0.001). Conclusion Combined locomotor training and aerobic exercise have increased muscle strength, as proven by increased handgrip strength. Combined locomotor training and aerobic exercise minimize fall risk and fracture in the elderly. Combined locomotor training and aerobic exercise are effective for the management of locomotive syndrome stage 1. Combined locomotor training and aerobic exercise reduce the GLFS-25 score.
Collapse
|
23
|
Mazo CE, Miranda ER, Shadiow J, Vesia M, Haus JM. High Intensity Acute Aerobic Exercise Elicits Alterations in Circulating and Skeletal Muscle Tissue Expression of Neuroprotective Exerkines. Brain Plast 2022; 8:5-18. [DOI: 10.3233/bpl-220137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2022] [Indexed: 11/15/2022] Open
Abstract
Background: Cathepsin B (CTSB) and brain derived neurotrophic factor (BDNF) are increased with aerobic exercise (AE) and skeletal muscle has been identified as a potential source of secretion. However, the intensity of AE and the potential for skeletal muscle contributions to circulating CTSB and BDNF have not been fully studied in humans. Objective: Determine the effects of AE intensity on circulating and skeletal muscle CTSB and BDNF expression profiles. Methods: Young healthy subjects (n = 16) completed treadmill-based AE consisting of VO2max and calorie-matched acute AE sessions at 40%, 65% and 80% VO2max. Fasting serum was obtained before and 30-minutes after each bout of exercise. Skeletal muscle biopsies (vastus lateralis) were taken before, 30-minutes and 3-hours after the 80% bout. Circulating CTSB and BDNF were assayed in serum. CTSB protein, BDNF protein and mRNA expression were measured in skeletal muscle tissue. Results: Serum CTSB increased by 20±7% (p = 0.02) and 30±18% (p = 0.04) after 80% and VO2max AE bouts, respectively. Serum BDNF showed a small non-significant increase (6±3%; p = 0.09) after VO2max. In skeletal muscle tissue, proCTSB increased 3 h-post AE (87±26%; p < 0.01) with no change in CTSB gene expression. Mature BDNF protein decreased (31±35%; p = 0.03) while mRNA expression increased (131±41%; p < 0.01) 3 h-post AE. Skeletal muscle fiber typing revealed that type IIa and IIx fibers display greater BDNF expression compared to type I (p = 0.02 and p < 0.01, respectively). Conclusions: High intensity AE elicits greater increases in circulating CTSB compared with lower intensities. Skeletal muscle protein and gene expression corroborate the potential role of skeletal muscle in generating and releasing neuroprotective exerkines into the circulation. NEW AND NOTEWORTHY: 1) CTSB is enriched in the circulation in an aerobic exercise intensity dependent manner. 2) Skeletal muscle tissue expresses both message and protein of CTSB and BDNF. 3) BDNF is highly expressed in glycolytic skeletal muscle fibers.
Collapse
Affiliation(s)
- Corey E. Mazo
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Edwin R. Miranda
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - James Shadiow
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Michael Vesia
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Jacob M. Haus
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
24
|
Bogacheva PO, Molchanova AI, Pravdivceva ES, Miteva AS, Balezina OP, Gaydukov AE. ProBDNF and Brain-Derived Neurotrophic Factor Prodomain Differently Modulate Acetylcholine Release in Regenerating and Mature Mouse Motor Synapses. Front Cell Neurosci 2022; 16:866802. [PMID: 35591942 PMCID: PMC9110780 DOI: 10.3389/fncel.2022.866802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022] Open
Abstract
The effects of brain-derived neurotrophic factor (BDNF) processing by-products (proBDNF and BDNF prodomain) on the activity of mouse neuromuscular junctions (NMJs) were studied in synapses formed during the reinnervation of extensor digitorum longus muscle (m. EDL) and mature synapses of the diaphragm. The parameters of spontaneous miniature endplate potentials (MEPPs) and evoked endplate potentials (EPPs) were analyzed in presence of each of the BDNF maturation products (both – 1 nM). In newly formed NMJs, proBDNF caused an increase in the resting membrane potential of muscle fibers and a decrease in the frequency of MEPPs, which was prevented by tertiapin-Q, a G-protein-coupled inwardly rectifying potassium channels (GIRK) blocker but not by p75 receptor signaling inhibitor TAT-Pep5. proBDNF had no effect on the parameters of EPPs. BDNF prodomain in newly formed synapses had effects different from those of proBDNF: it increased the amplitude of MEPPs, which was prevented by vesamicol, an inhibitor of vesicular acetylcholine (ACh) transporter; and reduced the quantal content of EPPs. In mature NMJs, proBDNF did not influence MEPPs parameters, but BDNF prodomain suppressed both spontaneous and evoked ACh release: decreased the frequency and amplitude of MEPPs, and the amplitude and quantal content of EPPs. This effect of the BDNF prodomain was prevented by blocking GIRK channels, by TAT-Pep5 or by Rho-associated protein kinase (ROCK) inhibitor Y-27632. At the same time, the BDNF prodomain did not show any inhibitory effects in diaphragm motor synapses of pannexin 1 knockout mice, which have impaired purinergic regulation of neuromuscular transmission. The data obtained suggest that there is a previously unknown mechanism for the acute suppression of spontaneous and evoked ACh release in mature motor synapses, which involves the activation of p75 receptors, ROCK and GIRK channels by BDNF prodomain and requires interaction with metabotropic purinoreceptors. In general, our results show that both the precursor of BDNF and the product of its maturation have predominantly inhibitory effects on spontaneous and evoked ACh release in newly formed or functionally mature neuromuscular junctions, which are mainly opposite to the effects of BDNF. The inhibitory influences of both proteins related to brain neurotrophin are mediated via GIRK channels of mouse NMJs.
Collapse
|
25
|
Involvement of the Voltage-Gated Calcium Channels L- P/Q- and N-Types in Synapse Elimination During Neuromuscular Junction Development. Mol Neurobiol 2022; 59:4044-4064. [PMID: 35474562 PMCID: PMC9167222 DOI: 10.1007/s12035-022-02818-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 03/22/2022] [Indexed: 10/26/2022]
Abstract
During the nervous system development, synapses are initially overproduced. In the neuromuscular junction (NMJ) however, competition between several motor nerve terminals and the synapses they made ends with the maturation of only one axon. The competitive signaling between axons is mediated by the differential activity-dependent release of the neurotransmitter ACh, co-transmitters, and neurotrophic factors. A multiple metabotropic receptor-driven downstream balance between PKA and PKC isoforms modulates the phosphorylation of targets involved in transmitter release and nerve terminal stability. Previously, we observed in the weakest endings on the polyinnervated NMJ that M1 mAChR receptors reduce ACh release through the PKC pathway coupled to an excess of Ca2+ inflow through P/Q- N- and L-type voltage-gated calcium channels (VGCC). This signaling would contribute to the elimination of this nerve terminal. Here, we investigate the involvement of the P/Q-, N-, and L-subtype channels in transgenic B6.Cg-Tg (Thy1-YFP)16-Jrs/J mice during synapse elimination. Then, the axon number and postsynaptic receptor cluster morphologic maturation were evaluated. The results show that both L- and P/Q-type VGCC (but not the N-type) are equally involved in synapse elimination. Their normal function favors supernumerary axonal loss by jointly enhancing intracellular calcium [Ca2+]i. The block of these VGCCs or [Ca2+]i i sequestration results in the same delay of axonal loss as the cPKCβI and nPKCε isoform block or PKA activation. The specific block of the muscle cell's contraction with μ-conotoxin GIIIB also delays synapse maturation, and thus, a retrograde influence from the postsynaptic site regulating the presynaptic CaV1.3 may contribute to the synapse elimination.
Collapse
|
26
|
Cefis M, Chaney R, Quirié A, Santini C, Marie C, Garnier P, Prigent-Tessier A. Endothelial cells are an important source of BDNF in rat skeletal muscle. Sci Rep 2022; 12:311. [PMID: 35013359 PMCID: PMC8748777 DOI: 10.1038/s41598-021-03740-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/09/2021] [Indexed: 12/27/2022] Open
Abstract
BDNF (brain-derived neurotrophic factor) is present in skeletal muscle, controlling muscular metabolism, strength and regeneration processes. However, there is no consensus on BDNF cellular source. Furthermore, while endothelial tissue expresses BDNF in large amount, whether endothelial cells inside muscle expressed BDNF has never been explored. The aim of the present study was to provide a comprehensive analysis of BDNF localization in rat skeletal muscle. Cellular localization of BDNF and activated Tropomyosin-related kinase B (TrkB) receptors was studied by immunohistochemical analysis on soleus (SOL) and gastrocnemius (GAS). BDNF and activated TrkB levels were also measured in muscle homogenates using Western blot analysis and/or Elisa tests. The results revealed BDNF immunostaining in all cell types examined with a prominent staining in endothelial cells and a stronger staining in type II than type I muscular fibers. Endothelial cells but not other cells displayed easily detectable activated TrkB receptor expression. Levels of BDNF and activated TrkB receptors were higher in SOL than GAS. In conclusion, endothelial cells are an important and still unexplored source of BDNF present in skeletal muscle. Endothelial BDNF expression likely explains why oxidative muscle exhibits higher BDNF levels than glycolytic muscle despite higher the BDNF expression by type II fibers.
Collapse
Affiliation(s)
- Marina Cefis
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Des Sciences de Santé, 21000, Dijon, France
| | - Remi Chaney
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Des Sciences de Santé, 21000, Dijon, France
| | - Aurore Quirié
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Des Sciences de Santé, 21000, Dijon, France
| | - Clélia Santini
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Des Sciences de Santé, 21000, Dijon, France
| | - Christine Marie
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Des Sciences de Santé, 21000, Dijon, France
| | - Philippe Garnier
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Des Sciences de Santé, 21000, Dijon, France
- Département Génie Biologique, IUT, 21000, Dijon, France
| | - Anne Prigent-Tessier
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR Des Sciences de Santé, 21000, Dijon, France.
- UFR Des Sciences de Santé, 7 boulevard Jeanne d'Arc, 21078, Dijon, France.
| |
Collapse
|
27
|
Moreira-Pais A, Ferreira R, Oliveira PA, Duarte JA. A neuromuscular perspective of sarcopenia pathogenesis: deciphering the signaling pathways involved. GeroScience 2022; 44:1199-1213. [PMID: 34981273 DOI: 10.1007/s11357-021-00510-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/26/2021] [Indexed: 12/18/2022] Open
Abstract
The escalation of life expectancy is accompanied by an increase in the prevalence of age-related conditions, such as sarcopenia. Sarcopenia, a muscle condition defined by low muscle strength, muscle quality or quantity, and physical performance, has a high prevalence among the elderly and is associated to increased mortality. The neuromuscular system has been emerging as a key contributor to sarcopenia pathogenesis. Indeed, the age-related degeneration of the neuromuscular junction (NMJ) function and structure may contribute to the loss of muscle strength and ultimately to the loss of muscle mass that characterize sarcopenia. The present mini-review discusses important signaling pathways involved in the function and maintenance of the NMJ, giving emphasis to the ones that might contribute to sarcopenia pathogenesis. Some conceivable biomarkers, such as C-terminal agrin fragment (CAF) and brain-derived neurotrophic factor (BDNF), and therapeutic targets, namely acetylcholine and calcitonin gene-related peptide (CGRP), can be retrieved, making way to future studies to validate their clinical use.
Collapse
Affiliation(s)
- Alexandra Moreira-Pais
- CIAFEL, Faculty of Sport, University of Porto, Dr. Plácido da Costa 91, 4200-450, Porto, Portugal. .,LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal. .,Centre for Research and Technology of Agro Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-Os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801, Vila Real, Portugal.
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Paula A Oliveira
- Centre for Research and Technology of Agro Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-Os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801, Vila Real, Portugal
| | - José A Duarte
- CIAFEL, Faculty of Sport, University of Porto, Dr. Plácido da Costa 91, 4200-450, Porto, Portugal.,TOXRUN - Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal
| |
Collapse
|
28
|
Cilleros-Mañé V, Just-Borràs L, Polishchuk A, Durán M, Tomàs M, Garcia N, Tomàs JM, Lanuza MA. M 1 and M 2 mAChRs activate PDK1 and regulate PKC βI and ε and the exocytotic apparatus at the NMJ. FASEB J 2021; 35:e21724. [PMID: 34133802 DOI: 10.1096/fj.202002213r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 05/07/2021] [Accepted: 05/24/2021] [Indexed: 01/14/2023]
Abstract
Neuromuscular junctions (NMJ) regulate cholinergic exocytosis through the M1 and M2 muscarinic acetylcholine autoreceptors (mAChR), involving the crosstalk between receptors and downstream pathways. Protein kinase C (PKC) regulates neurotransmission but how it associates with the mAChRs remains unknown. Here, we investigate whether mAChRs recruit the classical PKCβI and the novel PKCε isoforms and modulate their priming by PDK1, translocation and activity on neurosecretion targets. We show that each M1 and M2 mAChR activates the master kinase PDK1 and promotes a particular priming of the presynaptic PKCβI and ε isoforms. M1 recruits both primed-PKCs to the membrane and promotes Munc18-1, SNAP-25, and MARCKS phosphorylation. In contrast, M2 downregulates PKCε through a PKA-dependent pathway, which inhibits Munc18-1 synthesis and PKC phosphorylation. In summary, our results discover a co-dependent balance between muscarinic autoreceptors which orchestrates the presynaptic PKC and their action on ACh release SNARE-SM mechanism. Altogether, this molecular signaling explains previous functional studies at the NMJ and guide toward potential therapeutic targets.
Collapse
Affiliation(s)
- V Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - L Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - A Polishchuk
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M Durán
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - N Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - J M Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M A Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
29
|
Just-Borràs L, Cilleros-Mañé V, Hurtado E, Biondi O, Charbonnier F, Tomàs M, Garcia N, Tomàs J, Lanuza MA. Running and Swimming Differently Adapt the BDNF/TrkB Pathway to a Slow Molecular Pattern at the NMJ. Int J Mol Sci 2021; 22:4577. [PMID: 33925507 PMCID: PMC8123836 DOI: 10.3390/ijms22094577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/29/2022] Open
Abstract
Physical exercise improves motor control and related cognitive abilities and reinforces neuroprotective mechanisms in the nervous system. As peripheral nerves interact with skeletal muscles at the neuromuscular junction, modifications of this bidirectional communication by physical activity are positive to preserve this synapse as it increases quantal content and resistance to fatigue, acetylcholine receptors expansion, and myocytes' fast-to-slow functional transition. Here, we provide the intermediate step between physical activity and functional and morphological changes by analyzing the molecular adaptations in the skeletal muscle of the full BDNF/TrkB downstream signaling pathway, directly involved in acetylcholine release and synapse maintenance. After 45 days of training at different intensities, the BDNF/TrkB molecular phenotype of trained muscles from male B6SJLF1/J mice undergo a fast-to-slow transition without affecting motor neuron size. We provide further knowledge to understand how exercise induces muscle molecular adaptations towards a slower phenotype, resistant to prolonged trains of stimulation or activity that can be useful as therapeutic tools.
Collapse
Affiliation(s)
- Laia Just-Borràs
- Unitat d’Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, 43201 Reus, Spain; (L.J.-B.); (V.C.-M.); (E.H.); (M.T.); (N.G.)
| | - Víctor Cilleros-Mañé
- Unitat d’Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, 43201 Reus, Spain; (L.J.-B.); (V.C.-M.); (E.H.); (M.T.); (N.G.)
| | - Erica Hurtado
- Unitat d’Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, 43201 Reus, Spain; (L.J.-B.); (V.C.-M.); (E.H.); (M.T.); (N.G.)
| | - Olivier Biondi
- INSERM UMRS 1124, Université de Paris, CEDEX 06, F-75270 Paris, France; (O.B.); (F.C.)
| | - Frédéric Charbonnier
- INSERM UMRS 1124, Université de Paris, CEDEX 06, F-75270 Paris, France; (O.B.); (F.C.)
| | - Marta Tomàs
- Unitat d’Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, 43201 Reus, Spain; (L.J.-B.); (V.C.-M.); (E.H.); (M.T.); (N.G.)
| | - Neus Garcia
- Unitat d’Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, 43201 Reus, Spain; (L.J.-B.); (V.C.-M.); (E.H.); (M.T.); (N.G.)
| | - Josep Tomàs
- Unitat d’Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, 43201 Reus, Spain; (L.J.-B.); (V.C.-M.); (E.H.); (M.T.); (N.G.)
| | - Maria A. Lanuza
- Unitat d’Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, 43201 Reus, Spain; (L.J.-B.); (V.C.-M.); (E.H.); (M.T.); (N.G.)
| |
Collapse
|
30
|
Haddix SG, Rasband MN. Lose it to use it. J Cell Biol 2021; 220:e202102030. [PMID: 33734302 PMCID: PMC7980256 DOI: 10.1083/jcb.202102030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In this issue, Wang et al. (2021. J. Cell Biol.https://doi.org/10.1083/jcb.201911114) describe a phenomenon in which neuromuscular junction synapse elimination triggers myelination of terminal motor axon branches. They propose a mechanism initiated by synaptic pruning that depends on synaptic activity, cytoskeletal maturation, and the associated anterograde transport of trophic factors including Neuregulin 1-III.
Collapse
|
31
|
Effects of Physical Exercise on Neuroplasticity and Brain Function: A Systematic Review in Human and Animal Studies. Neural Plast 2021; 2020:8856621. [PMID: 33414823 PMCID: PMC7752270 DOI: 10.1155/2020/8856621] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/02/2020] [Accepted: 11/30/2020] [Indexed: 12/24/2022] Open
Abstract
Background Physical exercise (PE) has been associated with increase neuroplasticity, neurotrophic factors, and improvements in brain function. Objective To evaluate the effects of different PE protocols on neuroplasticity components and brain function in a human and animal model. Methods We conducted a systematic review process from November 2019 to January 2020 of the following databases: PubMed, ScienceDirect, SciELO, LILACS, and Scopus. A keyword combination referring to PE and neuroplasticity was included as part of a more thorough search process. From an initial number of 20,782 original articles, after reading the titles and abstracts, twenty-one original articles were included. Two investigators evaluated the abstract, the data of the study, the design, the sample size, the participant characteristics, and the PE protocol. Results PE increases neuroplasticity via neurotrophic factors (BDNF, GDNF, and NGF) and receptor (TrkB and P75NTR) production providing improvements in neuroplasticity, and cognitive function (learning and memory) in human and animal models. Conclusion PE was effective for increasing the production of neurotrophic factors, cell growth, and proliferation, as well as for improving brain functionality.
Collapse
|
32
|
Gong G, Chen H, Kam H, Chan G, Tang YX, Wu M, Tan H, Tse YC, Xu HX, Lee SMY. In Vivo Screening of Xanthones from Garcinia oligantha Identified Oliganthin H as a Novel Natural Inhibitor of Convulsions. JOURNAL OF NATURAL PRODUCTS 2020; 83:3706-3716. [PMID: 33296199 DOI: 10.1021/acs.jnatprod.0c00963] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Epilepsy is a chronic neurological disorder, characterized by recurrent, spontaneous, and transient seizures, and affects more than 70 million people worldwide. Although two dozen antiepileptic drugs (AEDs) are approved and available in the market, seizures remain poorly controlled in one-third of epileptic patients who are suffering from drug resistance or various adverse effects. Recently, the xanthone skeleton has been regarded as an attractive scaffold for the discovery and development of emerging anticonvulsants. We had isolated several dihydroxanthone derivatives previously, including oliganthin H, oliganthin I, and oliganthin N, whose structures were similar and delicately elucidated by spectrum analysis or X-ray crystallographic data, from extracts of leaves of Garcinia oligantha. These xanthone analogues were evaluated for anticonvulsant activity, and a novel xanthone, oliganthin H, has been identified as a sound and effective natural inhibitor of convulsions in zebrafish in vivo. A preliminary structure-activity relationship analysis on the relationship between structures of the xanthone analogues and their activities was also conducted. Oliganthin H significantly suppressed convulsant behavior and reduced to about 25% and 50% of PTZ-induced activity, in 12.5 and 25 μM treatment groups (P < 0.01 and 0.001), respectively. Meanwhile, it reduced seizure activity, velocity, seizure duration, and number of bursts in zebrafish larvae (P < 0.05). Pretreatment of oliganthin H significantly restored aberrant induction of gene expressions including npas4a, c-fos, pyya, and bdnf, as well as gabra1, gad1, glsa, and glula, upon PTZ treatment. In addition, in silico analysis revealed the stability of the oliganthin H-GABAA receptor complex and their detailed binding pattern. Therefore, direct interactions with the GABAA receptor and involvement of downstream GABA-glutamate pathways were possible mechanisms of the anticonvulsant action of oliganthin H. Our findings present the anticonvulsant activity of oliganthin H, provide a novel scaffold for further modifications, and highlight the xanthone skeleton as an attractive and reliable resource for the development of emerging AEDs.
Collapse
Affiliation(s)
- Guiyi Gong
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
- The Second Affiliated Hospital, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Hanbin Chen
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Hiotong Kam
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Ging Chan
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yue-Xun Tang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Man Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hongsheng Tan
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Yu-Chung Tse
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Department of Biology, Southern University of Science and Technology (SUSTech), Shenzhen, 518055, China
| | - Hong-Xi Xu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| |
Collapse
|
33
|
Le Blanc J, Fleury S, Boukhatem I, Bélanger JC, Welman M, Lordkipanidzé M. Platelets Selectively Regulate the Release of BDNF, But Not That of Its Precursor Protein, proBDNF. Front Immunol 2020; 11:575607. [PMID: 33324399 PMCID: PMC7723927 DOI: 10.3389/fimmu.2020.575607] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022] Open
Abstract
Background Brain-derived neurotrophic factor (BDNF) plays a role in synaptic plasticity and neuroprotection. BDNF has well-established pro-survival effects, whereas its precursor protein, proBDNF, induces apoptosis. Thus, it has been suggested that the proBDNF/BDNF ratio could be an indicator of neuronal health. Access to neurons is, understandably, limited. Because of their similarities, platelets have been put forward as a non-invasive biomarker of neuronal health; indeed, they store large quantities of BDNF and can release it into circulation upon activation, similarly to neurons. However, whether platelets also express the precursor proBDNF protein remains unknown. We therefore sought to characterize proBDNF levels in human platelets and plasma. Methods The presence of proBDNF was assessed by immunoblotting, cell fractionation, flow cytometry, and confocal microscopy in washed platelets from 10 healthy volunteers. Platelets from 20 independent healthy volunteers were activated with several classical agonists and the release of BDNF and proBDNF into plasma was quantified by ELISA. Results Platelets expressed detectable levels of proBDNF (21 ± 13 fmol/250 x 106 platelets). ProBDNF expression was mainly localized in the intracellular compartment. The proBDNF to BDNF molar ratio was ~1:5 in platelets and 10:1 in plasma. In stark contrast to the release of BDNF during platelet activation, intraplatelet and plasma concentrations of proBDNF remained stable following stimulation with classical platelet agonists, consistent with non-granular expression. Conclusions Platelets express both the mature and the precursor form of BDNF. Whether the intraplatelet proBDNF to BDNF ratio could be used as a non-invasive biomarker of cognitive health warrants further investigation.
Collapse
Affiliation(s)
- Jessica Le Blanc
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada.,Research Center, Montreal Heart Institute, Montréal, QC, Canada
| | - Samuel Fleury
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada.,Research Center, Montreal Heart Institute, Montréal, QC, Canada
| | - Imane Boukhatem
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada.,Research Center, Montreal Heart Institute, Montréal, QC, Canada
| | - Jean-Christophe Bélanger
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada.,Research Center, Montreal Heart Institute, Montréal, QC, Canada
| | - Mélanie Welman
- Research Center, Montreal Heart Institute, Montréal, QC, Canada
| | - Marie Lordkipanidzé
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada.,Research Center, Montreal Heart Institute, Montréal, QC, Canada
| |
Collapse
|
34
|
Molecular and neural adaptations to neuromuscular electrical stimulation; Implications for ageing muscle. Mech Ageing Dev 2020; 193:111402. [PMID: 33189759 PMCID: PMC7816160 DOI: 10.1016/j.mad.2020.111402] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/04/2020] [Accepted: 11/08/2020] [Indexed: 02/07/2023]
Abstract
Muscle atrophy and functional declines observed with advancing age can be minimized via various NMES protocols. Animal models have shown that NMES induces motor axon regeneration and promotes axonal outgrowth and fibre reinnervation. The activation of BDNF-trkB contributes to promotion of nerve growth and survival and mediates neuroplasticity. NMES is able to regulate muscle protein homeostasis and elevate oxidative enzyme activity.
One of the most notable effects of ageing is an accelerated decline of skeletal muscle mass and function, resulting in various undesirable outcomes such as falls, frailty, and all-cause mortality. The loss of muscle mass directly leads to functional deficits and can be explained by the combined effects of individual fibre atrophy and fibre loss. The gradual degradation of fibre atrophy is attributed to impaired muscle protein homeostasis, while muscle fibre loss is a result of denervation and motor unit (MU) remodelling. Neuromuscular electrical stimulation (NMES), a substitute for voluntary contractions, has been applied to reduce muscle mass and functional declines. However, the measurement of the effectiveness of NMES in terms of its mechanism of action on the peripheral motor nervous system and neuromuscular junction, and multiple molecular adaptations at the single fibre level is not well described. NMES mediates neuroplasticity and upregulates a number of neurotropic factors, manifested by increased axonal sprouting and newly formed neuromuscular junctions. Repeated involuntary contractions increase the activity levels of oxidative enzymes, increase fibre capillarisation and can influence fibre type conversion. Additionally, following NMES muscle protein synthesis is increased as well as functional capacity. This review will detail the neural, molecular, metabolic and functional adaptations to NMES in human and animal studies.
Collapse
|
35
|
Involvement of protein kinase C beta1-serotonin transporter system dysfunction in emotional behaviors in stressed mice. Neurochem Int 2020; 140:104826. [DOI: 10.1016/j.neuint.2020.104826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/27/2020] [Accepted: 08/07/2020] [Indexed: 12/17/2022]
|
36
|
Shen C, Zhou Y, Tang C, He C, Zuo Z. Developmental exposure to mepanipyrim induces locomotor hyperactivity in zebrafish (Danio rerio) larvae. CHEMOSPHERE 2020; 256:127106. [PMID: 32447115 DOI: 10.1016/j.chemosphere.2020.127106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 06/11/2023]
Abstract
Mepanipyrim is a widely used fungicide, and residues of mepanipyrim are frequently detected in commodities. However, the neurotoxicity and underlying mechanisms of mepanipyrim are still insufficiently understood. In this study, zebrafish embryos at 0.5-1.0 post-fertilization hours (hpf) were exposed to 0.1, 1, 10 and 100 μg/L mepanipyrim for 7 days. Our results showed that mepanipyrim could cause the locomotor hyperactivity and increase the concentration of γ-amino butyric acid (GABA) and the Na+/K+- and Ca2+-ATPase activities in zebrafish larvae. We have conducted the RNA-sequence and RT-qPCR to analyze the gene expressions. The mRNA expression levels of calcium/sodium ion conduction associated genes were observably up-regulated, demonstrating that mepanipyrim could enhance the cell energy metabolism, the synaptic transmission and skeletal muscle contraction, which were consistent with the locomotor hyperactivity. Meanwhile, exposure to mepanipyrim could significantly change the gene expression levels of gad1, bdnf, nlgn1, and type A and B GABA receptors in zebrafish larvae. This is the first study focusing on the underlying mechanisms of the neurotoxic effects that are induced by mepanipyrim.
Collapse
Affiliation(s)
- Chao Shen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yixi Zhou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chen Tang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China; State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, Fujian, 361005, China.
| |
Collapse
|
37
|
Bordoni B. The Five Diaphragms in Osteopathic Manipulative Medicine: Neurological Relationships, Part 2. Cureus 2020; 12:e8713. [PMID: 32699708 PMCID: PMC7372241 DOI: 10.7759/cureus.8713] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 06/20/2020] [Indexed: 02/06/2023] Open
Abstract
The main objective of the osteopath and that of osteopathic manipulative medicine (OMM) is to create space between the different tissues. The sliding capacity of the various tissue layers and between the different body components, up to the possibility of movement between cells is the salutogenic stimulus to allow the circulation of fluids, the biochemical exchange, and the adequate management of the multiple internal and external stimuli that perturb the body living. Movement is allowed by space and space is life. In this second part, the exposure of the anatomical neurological relationships of the five diaphragms continues, highlighting the relationships of the thoracic outlet, the respiratory diaphragm, and the pelvic floor. Finally, there will be clinical reflections to further corroborate the existence of the anatomical continuum and to lay the scientific foundations for an OMM approach to body diaphragms.
Collapse
Affiliation(s)
- Bruno Bordoni
- Physical Medicine and Rehabilitation, Foundation Don Carlo Gnocchi, Milan, ITA
| |
Collapse
|
38
|
Bogacheva P, Balezina O. Delayed increase of acetylcholine quantal size induced by the activity-dependent release of endogenous CGRP but not ATP in neuromuscular junctions. Synapse 2020; 74:e22175. [PMID: 32478912 DOI: 10.1002/syn.22175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/07/2020] [Accepted: 05/23/2020] [Indexed: 11/09/2022]
Abstract
In mouse motor synapses tetanic neuromuscular activity (30 Hz, 2 min) led to a delayed posttetanic potentiation of amplitude and duration of spontaneous miniature endplate potentials (MEPPs). Microelectrode recordings of MEPPs before and after nerve stimulation showed an increase in MEPP amplitude and time course by 30% and 15%, respectively, without changes in their frequency. Peak effect was detected 20 min after tetanic activity and progressively faded throughout the next 40 min of recording. The revealed potentiation of MEPPs was fully preserved in preparations from pannexin 1 knockout mice. It means, that myogenic ATP released via pannexin 1 channels from contracting muscle fibers is not likely to participate in the described phenomenon. But posttetanic potentiation of MEPPs was fully prevented by competitive antagonist of calcitonin gene-related peptide (CGRP) receptors CGRP8-37 , ryanodine receptors inhibitor ryanodine and by vesicular acetylcholine transporter inhibitor vesamicol. It is suggested that the combination of intensive synaptic and contractile activity in neuromuscular junctions is required to induce Ca2+ -dependent exocytosis of endogenous CGRP. The accumulation of CGRP in the synaptic cleft and its presynaptic activity may induce posttetanic potentiation of MEPP amplitude due to CGRP-stimulated acetylcholine loading into vesicles and subsequent increase of quantal size.
Collapse
Affiliation(s)
- Polina Bogacheva
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
| | - Olga Balezina
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
39
|
Takamori M. Myasthenia Gravis: From the Viewpoint of Pathogenicity Focusing on Acetylcholine Receptor Clustering, Trans-Synaptic Homeostasis and Synaptic Stability. Front Mol Neurosci 2020; 13:86. [PMID: 32547365 PMCID: PMC7272578 DOI: 10.3389/fnmol.2020.00086] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/28/2020] [Indexed: 12/18/2022] Open
Abstract
Myasthenia gravis (MG) is a disease of the postsynaptic neuromuscular junction (NMJ) where nicotinic acetylcholine (ACh) receptors (AChRs) are targeted by autoantibodies. Search for other pathogenic antigens has detected the antibodies against muscle-specific tyrosine kinase (MuSK) and low-density lipoprotein-related protein 4 (Lrp4), both causing pre- and post-synaptic impairments. Agrin is also suspected as a fourth pathogen. In a complex NMJ organization centering on MuSK: (1) the Wnt non-canonical pathway through the Wnt-Lrp4-MuSK cysteine-rich domain (CRD)-Dishevelled (Dvl, scaffold protein) signaling acts to form AChR prepatterning with axonal guidance; (2) the neural agrin-Lrp4-MuSK (Ig1/2 domains) signaling acts to form rapsyn-anchored AChR clusters at the innervated stage of muscle; (3) adaptor protein Dok-7 acts on MuSK activation for AChR clustering from “inside” and also on cytoskeleton to stabilize AChR clusters by the downstream effector Sorbs1/2; (4) the trans-synaptic retrograde signaling contributes to the presynaptic organization via: (i) Wnt-MuSK CRD-Dvl-β catenin-Slit 2 pathway; (ii) Lrp4; and (iii) laminins. The presynaptic Ca2+ homeostasis conditioning ACh release is modified by autoreceptors such as M1-type muscarinic AChR and A2A adenosine receptors. The post-synaptic structure is stabilized by: (i) laminin-network including the muscle-derived agrin; (ii) the extracellular matrix proteins (including collagen Q/perlecan and biglycan which link to MuSK Ig1 domain and CRD); and (iii) the dystrophin-associated glycoprotein complex. The study on MuSK ectodomains (Ig1/2 domains and CRD) recognized by antibodies suggested that the MuSK antibodies were pathologically heterogeneous due to their binding to multiple functional domains. Focussing one of the matrix proteins, biglycan which functions in the manner similar to collagen Q, our antibody assay showed the negative result in MG patients. However, the synaptic stability may be impaired by antibodies against MuSK ectodomains because of the linkage of biglycan with MuSK Ig1 domain and CRD. The pathogenic diversity of MG is discussed based on NMJ signaling molecules.
Collapse
|
40
|
Gaydukov AE, Dzhalagoniya IZ, Tarasova EO, Balezina OP. The Participation of Endocannabinoid Receptors in the Regulation of Spontaneous Synaptic Activity at Neuromuscular Junctions of Mice. BIOCHEMISTRY MOSCOW SUPPLEMENT SERIES A-MEMBRANE AND CELL BIOLOGY 2020. [DOI: 10.1134/s1990747819060059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
41
|
Cilleros-Mañé V, Just-Borràs L, Tomàs M, Garcia N, Tomàs JM, Lanuza MA. The M 2 muscarinic receptor, in association to M 1 , regulates the neuromuscular PKA molecular dynamics. FASEB J 2020; 34:4934-4955. [PMID: 32052889 DOI: 10.1096/fj.201902113r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/23/2019] [Accepted: 01/20/2020] [Indexed: 01/13/2023]
Abstract
Muscarinic acetylcholine receptor 1 subtype (M1 ) and muscarinic acetylcholine receptor 2 subtype (M2 ) presynaptic muscarinic receptor subtypes increase and decrease, respectively, neurotransmitter release at neuromuscular junctions. M2 involves protein kinase A (PKA), although the muscarinic regulation to form and inactivate the PKA holoenzyme is unknown. Here, we show that M2 signaling inhibits PKA by downregulating Cβ subunit, upregulating RIIα/β and liberating RIβ and RIIα to the cytosol. This promotes PKA holoenzyme formation and reduces the phosphorylation of the transmitter release target synaptosome-associated protein 25 and the gene regulator cAMP response element binding. Instead, M1 signaling, which is downregulated by M2 , opposes to M2 by recruiting R subunits to the membrane. The M1 and M2 reciprocal actions are performed through the anchoring protein A kinase anchor protein 150 as a common node. Interestingly, M2 modulation on protein expression needs M1 signaling. Altogether, these results describe the dynamics of PKA subunits upon M2 muscarinic signaling in basal and under presynaptic nerve activity, uncover a specific involvement of the M1 receptor and reveal the M1 /M2 balance to activate PKA to regulate neurotransmission. This provides a molecular mechanism to the PKA holoenzyme formation and inactivation which could be general to other synapses and cellular models.
Collapse
Affiliation(s)
- Víctor Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - Laia Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - Josep Maria Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - Maria Angel Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
42
|
Nakano I, Kinugawa S, Hori H, Fukushima A, Yokota T, Takada S, Kakutani N, Obata Y, Yamanashi K, Anzai T. Serum Brain-Derived Neurotrophic Factor Levels Are Associated with Skeletal Muscle Function but Not with Muscle Mass in Patients with Heart Failure. Int Heart J 2020; 61:96-102. [DOI: 10.1536/ihj.19-400] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Ippei Nakano
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Hiroaki Hori
- Department of Rehabilitation, Hokkaido University Hospital
| | - Arata Fukushima
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Takashi Yokota
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Shingo Takada
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Naoya Kakutani
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
- Research Fellow of the Japan Society for the Promotion of Science
| | - Yoshikuni Obata
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Katsuma Yamanashi
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| |
Collapse
|
43
|
Brain-derived neurotrophic factor-TrkB signaling in the medial prefrontal cortex plays a role in the anhedonia-like phenotype after spared nerve injury. Eur Arch Psychiatry Clin Neurosci 2020; 270:195-205. [PMID: 29882089 PMCID: PMC7036057 DOI: 10.1007/s00406-018-0909-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/31/2018] [Indexed: 12/14/2022]
Abstract
Although depressive symptoms including anhedonia (i.e., loss of pleasure) frequently accompany pain, little is known about the risk factors contributing to individual differences in pain-induced anhedonia. In this study, we examined if signaling of brain-derived neurotrophic factor (BDNF) and its receptor tropomyosin-receptor-kinase B (TrkB) contribute to individual differences in the development of neuropathic pain-induced anhedonia. Rats were randomly subjected to spared nerved ligation (SNI) or sham surgery. The SNI rats were divided into two groups based on the results of a sucrose preference test. Rats with anhedonia-like phenotype displayed lower tissue levels of BDNF in the medial prefrontal cortex (mPFC) compared with rats without anhedonia-like phenotype and sham-operated rats. In contrast, tissue levels of BDNF in the nucleus accumbens (NAc) of rats with an anhedonia-like phenotype were higher compared with those of rats without anhedonia-like phenotype and sham-operated rats. Furthermore, tissue levels of BDNF in the hippocampus, L2-5 spinal cord, muscle, and liver from both rats with or without anhedonia-like phenotype were lower compared with those of sham-operated rats. A single injection of 7,8-dihydroxyflavone (10 mg/kg; TrkB agonist), but not ANA-12 (0.5 mg/kg; TrkB antagonist), ameliorated reduced sucrose preference and reduced BDNF-TrkB signaling in the mPFC in the rats with anhedonia-like phenotype. These findings suggest that reduced BDNF-TrkB signaling in the mPFC might contribute to neuropathic pain-induced anhedonia, and that TrkB agonists could be potential therapeutic drugs for pain-induced anhedonia.
Collapse
|
44
|
Lee YI. Developmental neuromuscular synapse elimination: Activity-dependence and potential downstream effector mechanisms. Neurosci Lett 2019; 718:134724. [PMID: 31877335 DOI: 10.1016/j.neulet.2019.134724] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022]
Abstract
Synaptic connections initially formed during nervous system development undergo a significant transformation during nervous system maturation. Such maturation is essential for the proper architecture and function of the nervous system. Developmental synaptic transformation includes "synapse elimination," a process in which multiple immature presynaptic inputs converge at and compete for control of a common postsynaptic target. This developmental synaptic remodeling is best understood at mammalian neuromuscular junctions. It is well established that neuromuscular activity provides the impetus for the pruning of redundant motor axon inputs. Despite the dominant influence neuromuscular activity exerts on developmental synapse elimination, however, the downstream mechanisms of neuromuscular activity that affect synapse elimination remain poorly understood. Conversely, although several cellular and molecular effector mechanisms are known to impact synapse elimination, it is unclear whether they are modulated by neuromuscular activity. This review discusses how the motor neurons, synaptic glia and muscle fibers each contributes to the developmental phenomenon, and speculates how neuromuscular activity may modulate these contributions.
Collapse
Affiliation(s)
- Young Il Lee
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
45
|
KLF2 protects BV2 microglial cells against oxygen and glucose deprivation injury by modulating BDNF/TrkB pathway. Gene 2019; 735:144277. [PMID: 31821872 DOI: 10.1016/j.gene.2019.144277] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022]
Abstract
Cerebral ischemia injury is common in cerebral ischemic disease, and treatment options remain limited. Krueppel-like factor 2 (KLF2) is reported to negatively regulate inflammation in several ischemic diseases. Our study aimed to investigate the effects and underlying mechanism of KLF2 in BV2 microglial cells exposed to oxygen and glucose deprivation (OGD). We first found decreased KLF2 and toll-like receptor 2 (TLR2)/TLR4 in these cells. OGD also led to decrease in cell viability and increase in LDH release, apoptosis, the Bax/Bcl-2 ratio, and caspase3/9 expression, as well as production of inflammatory cytokines (e.g., TNFα, IL-1β and IL-6), reactive oxygen species (ROS), and TLR2/TLR4. To examine KLF2's effects on these OGD effects, we infected BV2 microglial cells with an ad-KLF2 or negative control vector, and we found that KLF2 reversed all of the effects of OGD exposure. Furthermore, KLF2 significantly increased levels of BDNF and TrkB in these cells, but these effects were blocked by K252a, a BDNF/TrkB inhibitor. K252a also decreased cell viability and increased apoptosis, inflammatory factors, ROS production, and TLR2/TLR4 expression in OGD-exposed BV2 cells that were treated with KLF2, were implying that K252a could reverse the effects of KLF2 on these cells. Taken together, our study results indicate that KLF2 may protect BV2 microglial cells against OGD injury by activating the BDNF/TrkB pathway.
Collapse
|
46
|
The Impact of Kinases in Amyotrophic Lateral Sclerosis at the Neuromuscular Synapse: Insights into BDNF/TrkB and PKC Signaling. Cells 2019; 8:cells8121578. [PMID: 31817487 PMCID: PMC6953086 DOI: 10.3390/cells8121578] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/19/2019] [Accepted: 12/03/2019] [Indexed: 12/14/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) promotes neuron survival in adulthood in the central nervous system. In the peripheral nervous system, BDNF is a contraction-inducible protein that, through its binding to tropomyosin-related kinase B receptor (TrkB), contributes to the retrograde neuroprotective control done by muscles, which is necessary for motor neuron function. BDNF/TrkB triggers downstream presynaptic pathways, involving protein kinase C, essential for synaptic function and maintenance. Undeniably, this reciprocally regulated system exemplifies the tight communication between nerve terminals and myocytes to promote synaptic function and reveals a new view about the complementary and essential role of pre and postsynaptic interplay in keeping the synapse healthy and strong. This signaling at the neuromuscular junction (NMJ) could establish new intervention targets across neuromuscular diseases characterized by deficits in presynaptic activity and muscle contractility and by the interruption of the connection between nervous and muscular tissues, such as amyotrophic lateral sclerosis (ALS). Indeed, exercise and other therapies that modulate kinases are effective at delaying ALS progression, preserving NMJs and maintaining motor function to increase the life quality of patients. Altogether, we review synaptic activity modulation of the BDNF/TrkB/PKC signaling to sustain NMJ function, its and other kinases’ disturbances in ALS and physical and molecular mechanisms to delay disease progression.
Collapse
|
47
|
Gultyaeva VV, Zinchenko MI, Uryumtsev DY, Krivoschekov SG, Aftanas LI. [Exercise for depression treatment. Physiological mechanisms]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:112-119. [PMID: 31464298 DOI: 10.17116/jnevro2019119071112] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This literature review considers meta-analyzes, systematic reviews and original research over the last decade addressing a comprehensive analysis of the antidepressant effect of targeted physical exercise and physical activity in general. Exercise is a promising non-pharmacological treatment for depression, showing effects that are comparable or may even exceed other first-line treatments of depression. The article introduces modern ideas about the mechanisms of depression and mechanisms of exercise effects on depression manifestations. The structures of the central nervous system, changing with the effective exercise-based treatment of depression, are indicated. Physical activity stimulates the secretion of growth factors, maintenance of angio-, synapto-, and neurogenesis. The regulation of antioxidant protection of neuronal mitochondria, a decrease in pro-inflammatory reactions and stress reactivity are also observed in response to regular exercise. Physical activity has a multimodal effect that stimulates biochemical pathways and restores neuronal structures disturbed in depression.
Collapse
Affiliation(s)
- V V Gultyaeva
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | - M I Zinchenko
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | - D Y Uryumtsev
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | - S G Krivoschekov
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | - L I Aftanas
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| |
Collapse
|
48
|
Opposed Actions of PKA Isozymes (RI and RII) and PKC Isoforms (cPKCβI and nPKCε) in Neuromuscular Developmental Synapse Elimination. Cells 2019; 8:cells8111304. [PMID: 31652775 PMCID: PMC6912401 DOI: 10.3390/cells8111304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/10/2019] [Accepted: 10/19/2019] [Indexed: 02/06/2023] Open
Abstract
Background: During neuromuscular junction (NMJ) development, synapses are produced in excess. By sensing the activity-dependent release of ACh, adenosine, and neurotrophins, presynaptic receptors prompt axonal competition and loss of the unnecessary axons. The receptor action is mediated by synergistic and antagonistic relations when they couple to downstream kinases (mainly protein kinases A and C (PKA and PKC)), which phosphorylate targets involved in axonal disconnection. Here, we directly investigated the involvement of PKA subunits and PKC isoforms in synapse elimination. Methods: Selective PKA and PKC peptide modulators were applied daily to the Levator auris longus (LAL) muscle surface of P5–P8 transgenic B6.Cg-Tg (Thy1-YFP) 16 Jrs/J (and also C57BL/6J) mice, and the number of axons and the postsynaptic receptor cluster morphology were evaluated in P9 NMJ. Results: PKA (PKA-I and PKA-II isozymes) acts at the pre- and postsynaptic sites to delay both axonal elimination and nAChR cluster differentiation, PKC activity promotes both axonal loss (a cPKCβI and nPKCε isoform action), and postsynaptic nAChR cluster maturation (a possible role for PKCθ). Moreover, PKC-induced changes in axon number indirectly influence postsynaptic maturation. Conclusions: PKC and PKA have opposed actions, which suggests that changes in the balance of these kinases may play a major role in the mechanism of developmental synapse elimination.
Collapse
|
49
|
Pérez V, Bermedo-Garcia F, Zelada D, Court FA, Pérez MÁ, Fuenzalida M, Ábrigo J, Cabello-Verrugio C, Moya-Alvarado G, Tapia JC, Valenzuela V, Hetz C, Bronfman FC, Henríquez JP. The p75 NTR neurotrophin receptor is required to organize the mature neuromuscular synapse by regulating synaptic vesicle availability. Acta Neuropathol Commun 2019; 7:147. [PMID: 31514753 PMCID: PMC6739937 DOI: 10.1186/s40478-019-0802-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/01/2019] [Indexed: 02/07/2023] Open
Abstract
The coordinated movement of organisms relies on efficient nerve-muscle communication at the neuromuscular junction. After peripheral nerve injury or neurodegeneration, motor neurons and Schwann cells increase the expression of the p75NTR pan-neurotrophin receptor. Even though p75NTR targeting has emerged as a promising therapeutic strategy to delay peripheral neuronal damage progression, the effects of long-term p75NTR inhibition at the mature neuromuscular junction have not been elucidated. We performed quantitative neuroanathomical analyses of the neuromuscular junction in p75NTR null mice by laser confocal and electron microscopy, which were complemented with electromyography, locomotor tests, and pharmacological intervention studies. Mature neuromuscular synapses of p75NTR null mice show impaired postsynaptic organization and ultrastructural complexity, which correlate with altered synaptic function at the levels of nerve activity-induced muscle responses, muscle fiber structure, force production, and locomotor performance. Our results on primary myotubes and denervated muscles indicate that muscle-derived p75NTR does not play a major role on postsynaptic organization. In turn, motor axon terminals of p75NTR null mice display a strong reduction in the number of synaptic vesicles and active zones. According to the observed pre and postsynaptic defects, pharmacological acetylcholinesterase inhibition rescued nerve-dependent muscle response and force production in p75NTR null mice. Our findings revealing that p75NTR is required to organize mature neuromuscular junctions contribute to a comprehensive view of the possible effects caused by therapeutic attempts to target p75NTR.
Collapse
Affiliation(s)
- Viviana Pérez
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Center for Advanced Microscopy (CMA BioBio), Universidad de Concepción, Concepción, Chile
| | - Francisca Bermedo-Garcia
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Center for Advanced Microscopy (CMA BioBio), Universidad de Concepción, Concepción, Chile
| | - Diego Zelada
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Center for Advanced Microscopy (CMA BioBio), Universidad de Concepción, Concepción, Chile
| | - Felipe A Court
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Miguel Ángel Pérez
- Laboratory of Neural Plasticity, Center for Neurobiology and Integrative Physiology, Faculty of Sciences, Institute of Physiology, Universidad de Valparaíso, Valparaíso, Chile
- Present Address: Health Sciences School, Universidad de Viña del Mar, Viña del Mar, Chile
| | - Marco Fuenzalida
- Laboratory of Neural Plasticity, Center for Neurobiology and Integrative Physiology, Faculty of Sciences, Institute of Physiology, Universidad de Valparaíso, Valparaíso, Chile
| | - Johanna Ábrigo
- Laboratory of Muscle Pathologies, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Millennium Institute on Immunology and Immunotherapy, Universidad Andrés Bello, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathologies, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Millennium Institute on Immunology and Immunotherapy, Universidad Andrés Bello, Santiago, Chile
| | - Guillermo Moya-Alvarado
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Carlos Tapia
- Department of Biomedical Sciences, Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| | - Vicente Valenzuela
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Francisca C Bronfman
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.
- Center for Aging and Regeneration (CARE), Institute of Biomedical Sciences (ICB), Faculty of Medicine and Faculty of Life Sciences, Universidad Andrés Bello, Santiago, Chile.
| | - Juan Pablo Henríquez
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Center for Advanced Microscopy (CMA BioBio), Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
50
|
Regulation of Acetylcholine Quantal Release by Coupled Thrombin/BDNF Signaling in Mouse Motor Synapses. Cells 2019; 8:cells8070762. [PMID: 31336670 PMCID: PMC6678150 DOI: 10.3390/cells8070762] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/13/2019] [Accepted: 07/22/2019] [Indexed: 12/23/2022] Open
Abstract
The aim of this study was to compare the acute effects of thrombin and brain-derived neurotrophic factor (BDNF) on spontaneous miniature endplate potentials (MEPPs) and multiquantal evoked endplate potentials (EPPs) in mouse neuromuscular junctions (NMJs) of m. diaphragma and m. EDL. Intracellular microelectrode recordings of MEPPs and EPPs were used to evaluate the changes in acetylcholine (ACh) release in mature and newly-formed mouse NMJs. Thrombin (1 nM) increased the amplitude of MEPPs and EPPs by 25–30% in mature and newly-formed NMJs. This effect was due to an enhanced loading of synaptic vesicles with ACh and increase of ACh quantal size, since it was fully prevented by blocking of vesicular ACh transporter. It was also prevented by tropomyosin-related kinase B (TrkB) receptors inhibitor ANA12. Exogenous BDNF (1 nM) mimicked thrombin effect and increased the amplitude of MEPPs and EPPs by 25–30%. It required involvement of protein kinase A (PKA) and mitogen-activated protein kinase (MEK1/2)-mediated pathway, but not phospholipase C (PLC). Blocking A2A adenosine receptors by ZM241385 abolished the effect of BDNF, whereas additional stimulation of A2A receptors by CGS21680 increased MEPP amplitudes, which was prevented by MEK1/2 inhibitor U0126. At mature NMJs, BDNF enhanced MEPPs frequency by 30–40%. This effect was selectively prevented by inhibition of PLC, but not PKA or MEK1/2. It is suggested that interrelated effects of thrombin/BDNF in mature and newly-formed NMJs are realized via enhancement of vesicular ACh transport and quantal size increase. BDNF-induced potentiation of synaptic transmission involves the functional coupling between A2A receptor-dependent active PKA and neurotrophin-triggered MAPK pathway, as well as PLC-dependent increase in frequency of MEPPs.
Collapse
|