Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell ; 187(10): 2465-2484.e22, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38701782

RESUMEN

Remyelination failure in diseases like multiple sclerosis (MS) was thought to involve suppressed maturation of oligodendrocyte precursors; however, oligodendrocytes are present in MS lesions yet lack myelin production. We found that oligodendrocytes in the lesions are epigenetically silenced. Developing a transgenic reporter labeling differentiated oligodendrocytes for phenotypic screening, we identified a small-molecule epigenetic-silencing-inhibitor (ESI1) that enhances myelin production and ensheathment. ESI1 promotes remyelination in animal models of demyelination and enables de novo myelinogenesis on regenerated CNS axons. ESI1 treatment lengthened myelin sheaths in human iPSC-derived organoids and augmented (re)myelination in aged mice while reversing age-related cognitive decline. Multi-omics revealed that ESI1 induces an active chromatin landscape that activates myelinogenic pathways and reprograms metabolism. Notably, ESI1 triggered nuclear condensate formation of master lipid-metabolic regulators SREBP1/2, concentrating transcriptional co-activators to drive lipid/cholesterol biosynthesis. Our study highlights the potential of targeting epigenetic silencing to enable CNS myelin regeneration in demyelinating diseases and aging.


Asunto(s)
Epigénesis Genética , Vaina de Mielina , Oligodendroglía , Remielinización , Animales , Vaina de Mielina/metabolismo , Humanos , Ratones , Remielinización/efectos de los fármacos , Oligodendroglía/metabolismo , Sistema Nervioso Central/metabolismo , Ratones Endogámicos C57BL , Rejuvenecimiento , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Organoides/metabolismo , Organoides/efectos de los fármacos , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/genética , Diferenciación Celular/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Masculino , Regeneración/efectos de los fármacos , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/genética , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología
2.
Cell Rep ; 43(5): 114129, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38640063

RESUMEN

The developing peripheral nervous and immune systems are functionally distinct from those of adults. These systems are vulnerable to early-life injury, which influences outcomes related to nociception following subsequent injury later in life (i.e., "neonatal nociceptive priming"). The underpinnings of this phenomenon are unclear, although previous work indicates that macrophages are trained by inflammation and injury. Our findings show that macrophages are both necessary and partially sufficient to drive neonatal nociceptive priming, possibly due to a long-lasting remodeling in chromatin structure. The p75 neurotrophic factor receptor is an important effector in regulating neonatal nociceptive priming through modulation of the inflammatory profile of rodent and human macrophages. This "pain memory" is long lasting in females and can be transferred to a naive host to alter sex-specific pain-related behaviors. This study reveals a mechanism by which acute, neonatal post-surgical pain drives a peripheral immune-related predisposition to persistent pain following a subsequent injury.

3.
JCI Insight ; 9(2)2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38258905

RESUMEN

Pain of unknown etiology is frequent in individuals with the tumor predisposition syndrome neurofibromatosis 1 (NF1), even when tumors are absent. Nerve Schwann cells (SCs) were recently shown to play roles in nociceptive processing, and we find that chemogenetic activation of SCs is sufficient to induce afferent and behavioral mechanical hypersensitivity in wild-type mice. In mouse models, animals showed afferent and behavioral hypersensitivity when SCs, but not neurons, lacked Nf1. Importantly, hypersensitivity corresponded with SC-specific upregulation of mRNA encoding glial cell line-derived neurotrophic factor (GDNF), independently of the presence of tumors. Neuropathic pain-like behaviors in the NF1 mice were inhibited by either chemogenetic silencing of SC calcium or by systemic delivery of GDNF-targeting antibodies. Together, these findings suggest that alterations in SCs directly modulate mechanical pain and suggest cell-specific treatment strategies to ameliorate pain in individuals with NF1.


Asunto(s)
Hipersensibilidad , Neuralgia , Neurofibromatosis 1 , Animales , Ratones , Neurofibromatosis 1/genética , Nocicepción , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Células de Schwann
4.
Brain Behav Immun ; 113: 156-165, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37442302

RESUMEN

A peripheral injury drives neuroimmune interactions at the level of the injury and throughout the neuraxis. Understanding these systems will be beneficial in the pursuit to target persistent pain that involves both neural and immune components. In this review, we discuss the impact of injury on the development of neuroimmune signaling, along with data that suggest a possible cellular immune memory. We also discuss the parallel effects of injury in the nervous system and immune related areas including bone marrow, lymph node and central nervous system-related cells. Finally, we relate these findings to patient populations and current research that evaluates human tissue.


Asunto(s)
Sistema Nervioso Central , Dolor , Humanos , Médula Ósea , Transducción de Señal , Neuroinmunomodulación
5.
bioRxiv ; 2023 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-37333316

RESUMEN

Repetitive ischemia with reperfusion (I/R) injury is a common cause of myalgia. I/R injuries occur in many conditions that differentially affect males and females including complex regional pain syndrome and fibromyalgia. Our preclinical studies have indicated that primary afferent sensitization and behavioral hypersensitivity due to I/R may be due to sex specific gene expression in the DRGs and distinct upregulation of growth factors and cytokines in the affected muscles. In order to determine how these unique gene expression programs may be established in a sex dependent manner in a model that more closely mimics clinical scenarios, we utilized a newly developed prolonged ischemic myalgia model in mice whereby animals experience repeated I/R injuries to the forelimb and compared behavioral results to unbiased and targeted screening strategies in male and female DRGs. Several distinct proteins were found to be differentially expressed in male and female DRGs, including AU-rich element RNA binding protein (AUF1), which is known to regulate gene expression. Nerve specific siRNA-mediated knockdown of AUF1 inhibited prolonged hypersensitivity in females only, while overexpression of AUF1 in male DRG neurons increased some pain-like responses. Further, AUF1 knockdown was able to specifically inhibit repeated I/R induced gene expression in females but not males. Data suggests that RNA binding proteins like AUF1 may underlie the sex specific effects on DRG gene expression that modulate behavioral hypersensitivity after repeated I/R injury. This study may aid in finding distinct receptor differences related to the evolution of acute to chronic ischemic muscle pain development between sexes.

6.
bioRxiv ; 2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-36824978

RESUMEN

The developing peripheral nervous and immune systems are functionally distinct from adults. These systems are vulnerable to early life injury, which influences outcomes related to nociception following subsequent injury later in life (neonatal nociceptive priming). The underpinnings of this phenomenon are largely unknown, although previous work indicates that macrophages are epigenetically trained by inflammation and injury. We found that macrophages are both necessary and partially sufficient to drive neonatal nociceptive priming possibly due to a long-lasting epigenetic remodeling. The p75 neurotrophic factor receptor (NTR) was an important effector in regulating neonatal nociceptive priming through modulation of the inflammatory profile of rodent and human macrophages. This pain memory was long lasting in females and could be transferred to a naive host to alter sex-specific pain-related behaviors. This study reveals a novel mechanism by which acute, neonatal post-surgical pain drives a peripheral immune-related predisposition to persistent pain following a subsequent injury.

7.
Can J Pain ; 6(2): 85-107, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35572362

RESUMEN

Background: Chronic postsurgical pain (CPSP) in children remains an important problem with no effective preventive or therapeutic strategies. Recently, genomic underpinnings explaining additional interindividual risk beyond psychological factors have been proposed. Aims: We present a comprehensive review of current preclinical and clinical evidence for genetic and epigenetic mechanisms relevant to pediatric CPSP. Methods: Narrative review. Results: Animal models are relevant to translational research for unraveling genomic mechanisms. For example, Cacng2, p2rx7, and bdnf mutant mice show altered mechanical hypersensitivity to injury, and variants of the same genes have been associated with CPSP susceptibility in humans; similarly, differential DNA methylation (H1SP) and miRNAs (miR-96/7a) have shown translational implications. Animal studies also suggest that crosstalk between neurons and immune cells may be involved in nociceptive priming observed in neonates. In children, differential DNA methylation in regulatory genomic regions enriching GABAergic, dopaminergic, and immune pathways, as well as polygenic risk scores for enhanced prediction of CPSP, have been described. Genome-wide studies in pediatric CPSP are scarce, but pathways identified by adult gene association studies point to potential common mechanisms. Conclusions: Bench-to-bedside genomics research in pediatric CPSP is currently limited. Reverse translational approaches, use of other -omics, and inclusion of pediatric/CPSP endophenotypes in large-scale biobanks may be potential solutions. Time of developmental vulnerability and longitudinal genomic changes after surgery warrant further investigation. Emergence of promising precision pain management strategies based on gene editing and epigenetic programing emphasize need for further research in pediatric CPSP-related genomics.


Contexte: La douleur chronique post-chirurgicale (DCPC) chez l'enfant reste un problème important pour lequel il n'y a pas de stratégies préventives ou thérapeutiques efficaces. Récemment, des fondements génomiques expliquant des risques interindividuels additionnels, au-delà des facteurs psychologiques, ont été proposés. Objectifs: Nous présentons une revue compléte des données probantes précliniques et cliniques actuelles pour les mécanismes génétiques et épigénétiques pertinents en matiére de DCPC pédiatrique. Méthodes: Revue narrative.Les modéles animaux sont pertinents pour la recherche translationnelle afin de déchiffrer les mécanismes génomiques. Par exemple, les souris mutantes Cacng2, p2rx7 et bdnf présentent une hypersensibilité mécanique altérée à des lésions et des variantes des mêmes génes ont été associées à la sensibilité à la DCPC chez l'humain; de même, la méthylation différentielle de l'ADN (H1SP) et les miARN (miR-96/7a) ont montré des implications translationnelles. Des études menées sur des animaux indiquent également que la diaphonie entre les neurones et les cellules immunitaires peut être impliquée dans l'amorçage nociceptif observé chez les nouveau-nés. Chez les enfants, la méthylation différentielle de l'ADN dans les régions génomiques régulatrices enrichissant les voies GABAergiques, dopaminergiques et immunitaires, ainsi que des scores de risque polygénique pour une prédiction améliorée de la PCSP, ont été décrits. Les études pangénomiques en matiére de DCPC pédiatrique sont rares, mais les voies identifiées par les études d'association de génes chez l'adulte indiquent de possibles mécanismes communs. Conclusions: La recherche en génomique du laboratoire au patient dans le cadre de la DCPC pédiatrique est actuellement limitée. Les approches translationnelles inversées, l'utilisation d'autres -omiques et l'inclusion d'endophénotypes pédiatriques/DCPC dans les biobanques à grande échelle peuvent être des solutions possibles. La durée de la vulnérabilité développementale et des changements génomiques longitudinaux aprés la chirurgie justifie des recherches plus approfondies. L'émergence de stratégies de précision prometteuses basées sur lé'dition de génes et la programmation épigénétique pour la prise en charge de la douleur font valoir la nécessité de poursuivre les recherches sur la génomique pédiatrique liée à la DCPC.

8.
eNeuro ; 9(2)2022.
Artículo en Inglés | MEDLINE | ID: mdl-35387844

RESUMEN

PEGPH20, a human recombinant hyaluronidase, has been proposed as a coadjutant to pancreatic cancer chemotherapy. In early trials, patients reported increased widespread muscle pain as the main adverse reaction to PEGPH20. To understand how PEGPH20 caused musculoskeletal pain, we systemically administered PEGPH20 to male mice and measured voluntary wheel activity and pain-related behaviors. These were paired with ex vivo electrophysiology of primary sensory neurons, whole DRG real-time PCR, and immunohistochemistry of hindpaw muscle. PEGPH20 induced significantly lower wheel running, compared with vehicle-treated animals, and decreased mechanical withdrawal thresholds 5 d after PEGPH20 injections. Chemo-sensory muscle afferents showed increased responses to noxious chemical stimulation of their receptive fields (RFs) in the PEGPH20-treated group. This was correlated with upregulation of the NGF receptor TrkA, the transient receptor potential vanilloid type 1 (TRPV1) channel and ATP-sensitive channel P2X3 in the DRG. Immunohistochemistry of hindpaw muscles revealed damage to the muscle architecture and extensive infiltration of the tissue by cells of the myelomonocytic lineage 3 d after PEGPH20 injection. Peripheral macrophage ablation in macrophage Fas-induced apoptosis (MaFIA) mice, however, did not prevent the decreased voluntary activity and instead caused even lower levels of running. These results suggest that disruption of hyaluronic acid (HA) within the muscle extracellular matrix (ECM) sensitizes chemo-nociceptive muscle afferents possibly leading to altered pain-like behaviors. Ablation experiments suggest macrophages are necessary for adequate recovery of voluntary activity after HA disruption. These data support a role for HA and macrophages in tissue integrity and muscle pain development in patients taking PEGPH20.


Asunto(s)
Ácido Hialurónico , Neoplasias Pancreáticas , Animales , Ganglios Espinales/fisiología , Humanos , Ácido Hialurónico/uso terapéutico , Masculino , Ratones , Actividad Motora , Músculo Esquelético , Mialgia , Neoplasias Pancreáticas/tratamiento farmacológico
9.
PM R ; 14(12): 1490-1496, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35077007

RESUMEN

Hyaluronan (HA) is a glycosaminoglycan that consists of single-chain polymers of disaccharide units of glucuronic acid and N-acetylglucosamine. It is a chief constituent of the extracellular matrix. About 27% of the total HA in the body is expressed in the skeleton and connective tissue, while 8% is expressed in muscles. In physiological conditions, HA functions as a lubricant and viscoelastic shock absorber. Additionally, HA is part of complex cellular signaling which modulates nociception and inflammation. This study aims to understand the role that HA plays in the musculoskeletal system, specifically in muscles and the surrounding fascia. This review is also intended to further understand HA homeostasis and the process of its synthesis, degradation, and clearance from the local tissue. The authors examined muscle pain and stiffness as pathological conditions associated with HA accumulation.


Asunto(s)
Matriz Extracelular , Ácido Hialurónico , Humanos , Ácido Hialurónico/metabolismo , Matriz Extracelular/metabolismo , Dolor , Homeostasis , Músculos/metabolismo
10.
Front Neurosci ; 15: 719735, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34566566

RESUMEN

Nociceptive nerve endings embedded in muscle tissue transduce peripheral noxious stimuli into an electrical signal [i.e., an action potential (AP)] to initiate pain sensations. A major contributor to nociception from the muscles is mechanosensation. However, due to the heterogeneity in the expression of proteins, such as ion channels, pumps, and exchangers, on muscle nociceptors, we currently do not know the relative contributions of different proteins and signaling molecules to the neuronal response due to mechanical stimuli. In this study, we employed an integrated approach combining a customized experimental study in mice with a computational model to identify key proteins that regulate mechanical nociception in muscles. First, using newly collected data from somatosensory recordings in mouse hindpaw muscles, we developed and then validated a computational model of a mechanosensitive mouse muscle nociceptor. Next, by performing global sensitivity analyses that simulated thousands of nociceptors, we identified three ion channels (among the 17 modeled transmembrane proteins and four endoplasmic reticulum proteins) as potential regulators of the nociceptor response to mechanical forces in both the innocuous and noxious range. Moreover, we found that simulating single knockouts of any of the three ion channels, delayed rectifier voltage-gated K+ channel (Kv1.1) or mechanosensitive channels Piezo2 or TRPA1, considerably altered the excitability of the nociceptor (i.e., each knockout increased or decreased the number of triggered APs compared to when all channels were present). These results suggest that altering expression of the gene encoding Kv1.1, Piezo2, or TRPA1 might regulate the response of mechanosensitive muscle nociceptors.

11.
J Neurosci ; 41(20): 4410-4427, 2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-33888610

RESUMEN

A number of cellular systems work in concert to modulate nociceptive processing in the periphery, but the mechanisms that regulate neonatal nociception may be distinct compared with adults. Our previous work indicated a relationship between neonatal hypersensitivity and growth hormone (GH) signaling. Here, we explored the peripheral mechanisms by which GH modulated neonatal nociception under normal and injury conditions (incision) in male and female mice. We found that GH receptor (GHr) signaling in primary afferents maintains a tonic inhibition of peripheral hypersensitivity. After injury, a macrophage dependent displacement of injury-site GH was found to modulate neuronal transcription at least in part via serum response factor (SRF) regulation. A single GH injection into the injured hindpaw muscle effectively restored available GH signaling to neurons and prevented acute pain-like behaviors, primary afferent sensitization, and neuronal gene expression changes. GH treatment also inhibited long-term somatosensory changes observed after repeated peripheral insult. Results may indicate a novel mechanism of neonatal nociception.SIGNIFICANCE STATEMENT Although it is noted that mechanisms of pain development in early life are unique compared with adults, little research focuses on neonatal-specific peripheral mechanisms of nociception. This gap is evident in the lack of specialized care for infants following an injury including surgeries. This report evaluates how distinct cellular systems in the periphery including the endocrine, immune and nervous systems work together to modulate neonatal-specific nociception. We uncovered a novel mechanism by which muscle injury induces a macrophage-dependent sequestration of peripheral growth hormone (GH) that effectively removes its normal tonic inhibition of neonatal nociceptors to promote acute pain-like behaviors. Results indicate a possible new strategy for treatment of neonatal postsurgical pain.


Asunto(s)
Hormona del Crecimiento/metabolismo , Macrófagos/metabolismo , Nocicepción/fisiología , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos , Femenino , Masculino , Ratones , Nociceptores/metabolismo , Receptores de Somatotropina/metabolismo
12.
Glia ; 69(8): 1837-1851, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33507559

RESUMEN

To facilitate analyses of purinergic signaling in peripheral nerve glia, we review recent literature and catalog purinergic receptor mRNA expression in cultured mouse Schwann cells (SCs). Purinergic signaling can decrease developmental SC proliferation, and promote SC differentiation. The purinergic receptors P2RY2 and P2RX7 are implicated in nerve development and in the ratio of Remak SCs to myelinating SCs in differentiated peripheral nerve. P2RY2, P2RX7, and other receptors are also implicated in peripheral neuropathies and SC tumors. In SC tumors lacking the tumor suppressor NF1, the SC pathway that suppresses SC growth through P2RY2-driven ß-arrestin-mediated AKT signaling is aberrant. SC-released purinergic agonists acting through SC and/or neuronal purinergic receptors activate pain responses. In all these settings, purinergic receptor activation can result in calcium-independent and calcium-dependent release of SC ATP and UDP, growth factors, and cytokines that may contribute to disease and nerve repair. Thus, current research suggests that purinergic agonists and/or antagonists might have the potential to modulate peripheral glia function in development and in disease.


Asunto(s)
Enfermedades del Sistema Nervioso Periférico , Células de Schwann , Animales , Ratones , Neuroglía/metabolismo , Enfermedades del Sistema Nervioso Periférico/metabolismo , Receptores Purinérgicos/metabolismo , Células de Schwann/metabolismo , Transducción de Señal/fisiología
13.
Mol Ther Methods Clin Dev ; 18: 84-97, 2020 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-32995353

RESUMEN

Gene therapy now provides a novel approach for treating inherited monogenetic disorders, including nuclear gene mutations associated with mitochondrial diseases. In this study, we have utilized a mouse model carrying a p.Arg389Gln mutation of the mitochondrial Ferredoxin Reductase gene (Fdxr) and treated them with neurotropic AAV-PHP.B vector loaded with the mouse Fdxr cDNA sequence. We then used immunofluorescence staining and western blot to test the transduction efficiency of this vector. Toluidine blue staining and electronic microscopy were also utilized to assess the morphology of optic and sciatic nerves, and the mitochondrial respiratory chain activity was determined as well. The AAV vector effectively transduced in the central nervous system and peripheral organs, and AAV-Fdxr treatment reversed almost all the symptoms of the mutants (Fdxr R389Q/R389Q ). This therapy also improved the electronic conductivity of the sciatic nerves, prevented optic atrophy, improved mobility, and restored mitochondrial complex function. Most notably, the sensory neuropathy, neurodegeneration, and chronic neuroinflammation in the brain were alleviated. Overall, we present the first demonstration of a potential definitive treatment that significantly improves optic and sciatic nerve atrophy, sensory neuropathy, and mitochondrial dysfunction in FDXR-related mitochondriopathy. Our study provides substantial support for the translation of AAV-based Fdxr gene therapy into clinical applications.

14.
Cell Death Dis ; 11(6): 423, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32499495

RESUMEN

The mitochondrial flavoprotein ferredoxin reductase (FDXR) is required for biogenesis of iron-sulfur clusters and for steroidogenesis. Iron-sulfur (Fe-S) clusters are ubiquitous cofactors essential to various cellular processes, and an increasing number of disorders are associated with disruptions in the synthesis of Fe-S clusters. Our previous studies have demonstrated that hypomorphic mutations in FDXR cause a novel mitochondriopathy and optic atrophy in humans and mice, attributed in part to reduced function of the electron transport chain (ETC) as well as elevated production of reactive oxygen species (ROS). Inflammation and peripheral neuropathy are also hallmarks of this disease. In this paper, we demonstrate that FDXR mutation leads to significant optic transport defects that are likely to underlie optic atrophy, a major clinical presentation in FDXR patients, as well as a neurodegenerative loss of cells in the central nervous system (CNS). Molecular analysis indicates that FDXR mutation also leads to mitochondrial iron overload and an associated depolarization of the mitochondrial membrane, further supporting the hypothesis that FDXR mutations cause neurodegeneration by affecting FDXR's critical role in iron homeostasis.


Asunto(s)
Proteínas Mitocondriales/genética , Enfermedades del Nervio Óptico/genética , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Animales , Atrofia , Axones/patología , Transporte Biológico , Línea Celular , Marcha , Humanos , Hierro/metabolismo , Potencial de la Membrana Mitocondrial , Ratones Mutantes , Mutación/genética , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Enfermedades del Nervio Óptico/patología , Enfermedades del Nervio Óptico/fisiopatología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Neuronas Retinianas/metabolismo , Neuronas Retinianas/patología
15.
Hum Mol Genet ; 29(4): 649-661, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-31943007

RESUMEN

Mitochondrial disorders are the result of nuclear and mitochondrial DNA mutations that affect multiple organs, with the central and peripheral nervous system often affected. Currently, there is no cure for mitochondrial disorders. Currently, gene therapy offers a novel approach for treating monogenetic disorders, including nuclear genes associated with mitochondrial disorders. We utilized a mouse model carrying a knockout of the mitochondrial fusion-fission-related gene solute carrier family 25 member 46 (Slc25a46) and treated them with neurotrophic AAV-PHP.B vector carrying the mouse Slc25a46 coding sequence. Thereafter, we used immunofluorescence staining and western blot to test the transduction efficiency of this vector. Toluidine blue staining and electronic microscopy were utilized to assess the morphology of optic and sciatic nerves following treatment, and the morphology and respiratory chain activity of mitochondria within these tissues were determined as well. The adeno-associated virus (AAV) vector effectively transduced in the cerebrum, cerebellum, heart, liver and sciatic nerves. AAV-Slc25a46 treatment was able to rescue the premature death in the mutant mice (Slc25a46-/-). The treatment-improved electronic conductivity of the peripheral nerves increased mobility and restored mitochondrial complex activities. Most notably, mitochondrial morphology inside the tissues of both the central and peripheral nervous systems was normalized, and the neurodegeneration, chronic neuroinflammation and loss of Purkinje cell dendrites observed within the mutant mice were alleviated. Overall, our study shows that AAV-PHP.B's neurotrophic properties are plausible for treating conditions where the central nervous system is affected, such as many mitochondrial diseases, and that AAV-Slc25a46 could be a novel approach for treating SLC25A46-related mitochondrial disorders.


Asunto(s)
Ataxia/prevención & control , Enfermedades del Sistema Nervioso Central/prevención & control , Dependovirus/genética , Terapia Genética , Vectores Genéticos/administración & dosificación , Enfermedades Mitocondriales/prevención & control , Proteínas de Transporte de Fosfato/fisiología , Animales , Ataxia/genética , Ataxia/patología , Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología
16.
Proc Natl Acad Sci U S A ; 117(1): 698-707, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31848242

RESUMEN

Group III/IV muscle afferents transduce nociceptive signals and modulate exercise pressor reflexes (EPRs). However, the mechanisms governing afferent responsiveness to dually modulate these processes are not well characterized. We and others have shown that ischemic injury can induce both nociception-related behaviors and exacerbated EPRs in the same mice. This correlated with primary muscle afferent sensitization and increased expression of glial cell line-derived neurotrophic factor (GDNF) in injured muscle and increased expression of GDNF family receptor α1 (GFRα1) in dorsal root ganglia (DRG). Here, we report that increased GDNF/GFRα1 signaling to sensory neurons from ischemia/reperfusion-affected muscle directly modulated nociceptive-like behaviors and increased exercise-mediated reflexes and group III/IV muscle afferent sensitization. This appeared to have taken effect through increased cyclic adenosine monophosphate (cAMP) response element binding (CREB)/CREB binding protein-mediated expression of the purinergic receptor P2X5 in the DRGs. Muscle GDNF signaling to neurons may, therefore, play an important dual role in nociception and sympathetic reflexes and could provide a therapeutic target for treating complications from ischemic injuries.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Mialgia/etiología , Nocicepción/fisiología , Reflejo/fisiología , Daño por Reperfusión/patología , Animales , Proteína de Unión a CREB/metabolismo , Sistema Cardiovascular/inervación , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Ejercicio Físico/fisiología , Ganglios Espinales/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Frecuencia Cardíaca/fisiología , Humanos , Masculino , Ratones , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/inervación , Músculo Esquelético/metabolismo , Mialgia/patología , Neuronas Aferentes/fisiología , Receptores Purinérgicos P2X5/metabolismo , Daño por Reperfusión/complicaciones , Transducción de Señal/fisiología
18.
Curr Opin Physiol ; 11: 1-6, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31245656

RESUMEN

Clinical conditions resulting in musculoskeletal pain show important sex differences in both prevalence and degree of functional disability. The underlying mechanisms for these distinctions in pain manifestation are not fully known. However, recent preclinical studies have shown at the primary afferent level that males and females present fundamental differences in their peripheral response properties and injury-related gene expression patterns that may underlie observed afferent sensitization. At the spinal cord level, studies in various models of pain suggest important roles for the immune system, glutamate signaling and hormones in modulating sex differences. While preclinical studies have been able to characterize some of the basic underlying molecular mechanisms of sex differences in muscle pain, human studies have relied mainly on functional brain imaging studies to explain differences. Further complicating our understanding of how sex influences muscle pain is the notion that the type of injury sustained, or clinical condition may differentially activate distinct mechanisms of muscle pain development in males versus females. More research is necessary to better understand how the sexes differ in their perception of muscle pain. This review highlights recent advances in both human and animal studies of sex differences in muscle pain.

19.
PLoS One ; 14(5): e0216527, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31107888

RESUMEN

In addition to large plexiform neurofibromas (pNF), NF1 patients are frequently disfigured by cutaneous neurofibromas (cNF) and are often afflicted with chronic pain and itch even from seemingly normal skin areas. Both pNFs and cNF consist primarily of benign hyperproliferating nonmyelinating Schwann cells (nSC). While pNF clearly arise within deep nerves and plexuses, the role of cutaneous innervation in the origin of cNF and in chronic itch and pain is unknown. First, we conducted a comprehensive, multi-molecular, immunofluorescence (IF) analyses on 3mm punch biopsies from three separate locations in normal appearing, cNF-free skin in 19 NF1 patients and skin of 16 normal subjects. At least one biopsy in 17 NF1 patients had previously undescribed micro-lesions consisting of a small, dense cluster of nonpeptidergic C-fiber endings and the affiliated nSC consistently adjoining adnexal structures-dermal papillae, hair follicles, sweat glands, sweat ducts, and arterioles-where C-fiber endings normally terminate. Similar micro-lesions were detected in hind paw skin of mice with conditionally-induced SC Nf1-/- mutations. Hypothesizing that these microlesions were pre-cNF origins of cNF, we subsequently analyzed numerous overt, small cNF (s-cNF, 3-6 mm) and discovered that each had an adnexal structure at the epicenter of vastly increased nonpeptidergic C-fiber terminals, accompanied by excessive nSC. The IF and functional genomics assays indicated that neurturin (NTRN) and artemin (ARTN) signaling through cRET kinase and GFRα2 and GFRα3 co-receptors on the aberrant C-fiber endings and nSC may mutually promote the onset of pre-cNF and their evolution to s-cNF. Moreover, TrpA1 and TrpV1 receptors may, respectively, mediate symptoms of chronic itch and pain. These newly discovered molecular characteristics might be targeted to suppress the development of cNF and to treat chronic itch and pain symptoms in NF1 patients.


Asunto(s)
Fibras Nerviosas Amielínicas/metabolismo , Neurofibroma Plexiforme/patología , Neurofibromatosis 1/patología , Células de Schwann/metabolismo , Neoplasias Cutáneas/patología , Anciano , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fibras Nerviosas Amielínicas/patología , Proteínas del Tejido Nervioso/metabolismo , Neurofibroma Plexiforme/metabolismo , Neurofibromatosis 1/inmunología , Neurturina/metabolismo , Células de Schwann/patología , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Adulto Joven
20.
IBRO Rep ; 6: 111-121, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30815617

RESUMEN

Injury during early postnatal life causes acute alterations in afferent function and DRG gene expression, which in addition to producing short-term sensitivity has the potential to influence nociceptive responses in adulthood. We recently discovered that growth hormone (GH) is a key regulator of afferent sensitization and pain-related behaviors during developmental inflammation of the skin. Peripheral injury caused a significant reduction in cutaneous GH levels, which corresponded with the observed hypersensitivity. However, it has yet to be determined whether GH deficiency (GHD) is sufficient to drive peripheral sensitization in uninjured animals. Here, we found that systemic GHD, induced by knockout of the GH release hormone receptor (GHRHr), was able to induce behavioral and afferent hypersensitivity to peripheral stimuli specifically during early developmental stages. GHD also produced an upregulation of many receptors and channels linked to nociceptive processing in the DRGs at these early postnatal ages (P7 and P14). Surprisingly, P21 GHRHr knockouts also displayed significant alterations in DRG gene expression even though behavioral and afferent hypersensitivity resolved. These data support previous findings that GH is a key modulator of neonatal hypersensitivity. Results may provide insight into whether GH treatment may be a therapeutic strategy for pediatric pain.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...