Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Prog Community Health Partnersh ; 17(2): 347-351, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37462563

RESUMEN

BACKGROUND: Community-academic partnerships are increasingly used in interventions to address health care disparities. Little is known about motivations and perceptions of participating community members. OBJECTIVES: To elicit community members' perspectives of involvement in a community-academic partnership to address implicit bias in health care. METHODS: With our partnering community organizer, we conducted one-on-one semistructured interviews and a follow-up group interview with participating community members to solicit experiences about involvement in an National Institutes of Health-funded clinician training; responses were organized using content analysis. RESULTS: Community members revealed that their participation was motivated by trust in our community organizer; they derived personal pride from participation in clinician training; the power differential between community members and clinicians in the training environment needed to be levelled. Our community organizer noted that the benefits of community-academic partnerships propagate to the larger community via community members' experiences. CONCLUSIONS: Community members note trust, pride, and power as important elements in community-academic partnership.


Asunto(s)
Sesgo Implícito , Investigación Participativa Basada en la Comunidad , Humanos , Universidades , Relaciones Comunidad-Institución , Conducta Cooperativa
2.
Mol Neurobiol ; 57(6): 2521-2538, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32198698

RESUMEN

Mitochondrial dysfunction is implicated in a variety of neurodegenerative diseases of the nervous system. Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) is a regulator of mitochondrial function in multiple cell types. In sensory neurons, AMP-activated protein kinase (AMPK) augments PGC-1α activity and this pathway is depressed in diabetes leading to mitochondrial dysfunction and neurodegeneration. Antimuscarinic drugs targeting the muscarinic acetylcholine type 1 receptor (M1R) prevent/reverse neurodegeneration by inducing nerve regeneration in rodent models of diabetes and chemotherapy-induced peripheral neuropathy (CIPN). Ca2+/calmodulin-dependent protein kinase kinase ß (CaMKKß) is an upstream regulator of AMPK activity. We hypothesized that antimuscarinic drugs modulate CaMKKß to enhance activity of AMPK, and PGC-1α, increase mitochondrial function and thus protect from neurodegeneration. We used the specific M1R antagonist muscarinic toxin 7 (MT7) to manipulate muscarinic signaling in the dorsal root ganglia (DRG) neurons of normal rats or rats with streptozotocin-induced diabetes. DRG neurons treated with MT7 (100 nM) or a selective muscarinic antagonist, pirenzepine (1 µM), for 24 h showed increased neurite outgrowth that was blocked by the CaMKK inhibitor STO-609 (1 µM) or short hairpin RNA to CaMKKß. MT7 enhanced AMPK phosphorylation which was blocked by STO-609 (1 µM). PGC-1α reporter activity was augmented up to 2-fold (p < 0.05) by MT7 and blocked by STO-609. Mitochondrial maximal respiration and spare respiratory capacity were elevated after 3 h of exposure to MT7 (p < 0.05). Diabetes and CIPN induced a significant (p < 0.05) decrease in corneal nerve density which was corrected by topical delivery of MT7. We reveal a novel M1R-modulated, CaMKKß-dependent pathway in neurons that represents a therapeutic target to enhance nerve repair in two of the most common forms of peripheral neuropathy.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Venenos Elapídicos/farmacología , Mitocondrias/efectos de los fármacos , Degeneración Nerviosa/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Diabetes Mellitus Experimental/metabolismo , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Mitocondrias/metabolismo , Antagonistas Muscarínicos/farmacología , Proyección Neuronal/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Fosforilación/efectos de los fármacos , Pirenzepina/farmacología , Ratas , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo
3.
J Clin Invest ; 127(2): 608-622, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-28094765

RESUMEN

Sensory neurons have the capacity to produce, release, and respond to acetylcholine (ACh), but the functional role of cholinergic systems in adult mammalian peripheral sensory nerves has not been established. Here, we have reported that neurite outgrowth from adult sensory neurons that were maintained under subsaturating neurotrophic factor conditions operates under cholinergic constraint that is mediated by muscarinic receptor-dependent regulation of mitochondrial function via AMPK. Sensory neurons from mice lacking the muscarinic ACh type 1 receptor (M1R) exhibited enhanced neurite outgrowth, confirming the role of M1R in tonic suppression of axonal plasticity. M1R-deficient mice made diabetic with streptozotocin were protected from physiological and structural indices of sensory neuropathy. Pharmacological blockade of M1R using specific or selective antagonists, pirenzepine, VU0255035, or muscarinic toxin 7 (MT7) activated AMPK and overcame diabetes-induced mitochondrial dysfunction in vitro and in vivo. These antimuscarinic drugs prevented or reversed indices of peripheral neuropathy, such as depletion of sensory nerve terminals, thermal hypoalgesia, and nerve conduction slowing in diverse rodent models of diabetes. Pirenzepine and MT7 also prevented peripheral neuropathy induced by the chemotherapeutic agents dichloroacetate and paclitaxel or HIV envelope protein gp120. As a variety of antimuscarinic drugs are approved for clinical use against other conditions, prompt translation of this therapeutic approach to clinical trials is feasible.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Neuropatías Diabéticas/tratamiento farmacológico , Hiperalgesia/tratamiento farmacológico , Antagonistas Muscarínicos/farmacología , Receptor Muscarínico M1/antagonistas & inhibidores , Células Receptoras Sensoriales/metabolismo , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Neuropatías Diabéticas/genética , Neuropatías Diabéticas/metabolismo , Neuropatías Diabéticas/patología , Hiperalgesia/genética , Hiperalgesia/metabolismo , Masculino , Ratones , Ratones Mutantes , Mitocondrias/metabolismo , Mitocondrias/patología , Neuritas/metabolismo , Neuritas/patología , Ratas , Receptor Muscarínico M1/genética , Células Receptoras Sensoriales/patología
4.
Cell Mol Neurobiol ; 34(5): 643-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24682898

RESUMEN

Mitochondrial dysfunction occurs in sensory neurons and contributes to diabetic neuropathy. Ciliary neurotrophic factor (CNTF) stimulates axon regeneration in type 1 diabetic rodents and prevents deficits in axonal caliber, nerve conduction, and thermal sensation. We tested the hypothesis that CNTF enhances sensory neuron function in diabetes through JAK/STAT (Janus kinase/signal transducers and activators of transcription) signaling to normalize impaired mitochondrial bioenergetics. The effect of CNTF on gene expression and neurite outgrowth of cultured adult dorsal root ganglia (DRG) sensory neurons derived from control and streptozotocin (STZ)-induced diabetic rodents was quantified. Polarization status and bioenergetics profile of mitochondria from cultured sensory neurons were determined. CNTF treatment prevented reduced STAT3 phosphorylation (Tyr 705) in DRG of STZ-diabetic mice and also enhanced STAT3 phosphorylation in rat DRG cultures. CNTF normalized polarization status of the mitochondrial inner membrane and corrected the aberrant oligomycin-induced mitochondrial hyperpolarization in axons of diabetic neurons. The mitochondrial bioenergetics profile demonstrated that spare respiratory capacity and respiratory control ratio were significantly depressed in sensory neurons cultured from STZ-diabetic rats and were corrected by acute CNTF treatment. The positive effects of CNTF on neuronal mitochondrial function were significantly inhibited by the specific JAK inhibitor, AG490. Neurite outgrowth of sensory neurons from age-matched control and STZ-induced diabetic rats was elevated by CNTF and blocked by AG490. We propose that CNTF's ability to enhance axon regeneration and protect from fiber degeneration in diabetes is associated with its targeting of mitochondrial function and improvement of cellular bioenergetics, in part, through JAK/STAT signaling.


Asunto(s)
Factor Neurotrófico Ciliar/farmacología , Diabetes Mellitus Experimental/metabolismo , Metabolismo Energético/fisiología , Quinasas Janus/metabolismo , Factor de Transcripción STAT3/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Células Cultivadas , Factor Neurotrófico Ciliar/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Metabolismo Energético/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
5.
Mol Brain ; 6: 45, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24152426

RESUMEN

BACKGROUND: A luminex-based screen of cytokine expression in dorsal root ganglia (DRG) and nerve of type 1 diabetic rodents revealed interleukin-1 (IL-1α) and IL-1ß to be significantly depressed. We, therefore, tested the hypothesis that impaired IL-1α and IL-1ß expression in DRG may contribute to aberrant axon regeneration and plasticity seen in diabetic sensory neuropathy. In addition, we determined if these cytokines could optimize mitochondrial bioenergetics since mitochondrial dysfunction is a key etiological factor in diabetic neuropathy. RESULTS: Cytokines IL-1α and IL-1ß were reduced 2-fold (p<0.05) in DRG and/or nerve of 2 and 5 month streptozotocin (STZ)-diabetic rats. IL-2 and IL-10 were unchanged. IL-1α and IL-1ß induced similar 2 to 3-fold increases in neurite outgrowth in cultures derived from control or diabetic rats (p<0.05). STAT3 phosphorylation on Tyr705 or Ser727 was depressed in DRG from STZ-diabetic mice and treatment of cultures derived from STZ-diabetic rats with IL-1ß for 30 min raised phosphorylation of STAT3 on Tyr705 and Ser727 by 1.5 to 2-fold (p<0.05). shRNA-based or AG490 inhibition of STAT3 activity or shRNA blockade of endogenous IL-1ß expression completely blocked neurite outgrowth. Cultured neurons derived from STZ-diabetic mice were treated for 24 hr with IL-1ß and maximal oxygen consumption rate and spare respiratory capacity, both key measures of bioenergetic fidelity that were depressed in diabetic compared with control neurons, were enhanced 2-fold. This effect was blocked by AG490. CONCLUSIONS: Endogenous synthesis of IL-1ß is diminished in nerve tissue in type 1 diabetes and we propose this defect triggers reduced STAT3 signaling and mitochondrial function leading to sup-optimal axonal regeneration and plasticity.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Metabolismo Energético , Interleucina-1beta/metabolismo , Quinasas Janus/metabolismo , Mitocondrias/metabolismo , Neuritas/metabolismo , Factor de Transcripción STAT3/metabolismo , Envejecimiento/metabolismo , Animales , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/patología , Metabolismo Energético/efectos de los fármacos , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Inmunohistoquímica , Interleucina-1alfa/metabolismo , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Neuritas/efectos de los fármacos , Neuritas/enzimología , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Ratas , Ratas Sprague-Dawley , Nervio Ciático/efectos de los fármacos , Nervio Ciático/metabolismo , Nervio Ciático/patología , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Transducción de Señal/efectos de los fármacos , Estreptozocina , Tirfostinos/farmacología
6.
Exp Neurol ; 249: 149-59, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24029001

RESUMEN

BACKGROUND: The receptor for advanced glycation end-products (RAGE) is implicated in neuronal differentiation during embryogenesis and in regulation of peripheral nerve regeneration. However, the role of RAGE ligands and the signaling pathways utilized by activated RAGE in mediating axon regeneration in adult neurons remain unknown. We tested the hypothesis that RAGE signaling modulated neurotrophin-induced neurite outgrowth in cultured adult sensory neurons. RESULTS: Dorsal root ganglia (DRG) neurons from adult rats in vitro were exposed to specific RAGE ligands, signal transduction inhibitors and function blocking anti-RAGE IgG to assess their impact on neurite outgrowth. RAGE ligands including human glycated albumin (HGA), S100 calcium binding protein (S100B) and high mobility group 1 protein (HMGB1; alternatively termed amphoterin) in the presence of neurotrophins elevated neurite outgrowth 2-fold (p<0.05). shRNA to RAGE or anti-RAGE IgG blockade of RAGE inhibited neurite outgrowth by 40-90% (p<0.05). Western blotting and gene reporter analysis showed RAGE ligands activated NF-κB, JAK-STAT and ERK pathways. RAGE ligand induction of neurite outgrowth was blocked by inhibition of NF-κB, JAK-STAT or ERK pathways revealing the necessity for combined activation for optimal growth. RAGE ligands rapidly elevated NF-κB p65 expression in the cytoplasm while triggering translocation of NF-κB p50 to the nucleus. shRNA blockade of p50 demonstrated that translocation of p50 to the nucleus was implicated in driving axonal outgrowth. CONCLUSIONS: RAGE signaling is a complex mediator of neurotrophin-dependent neurite outgrowth, operating through divergent but partly inter-dependent pathways.


Asunto(s)
Neuritas/fisiología , Receptores Inmunológicos/fisiología , Células Receptoras Sensoriales/fisiología , Transducción de Señal/fisiología , Envejecimiento/fisiología , Animales , Células Cultivadas , Ganglios Espinales/citología , Ganglios Espinales/fisiología , Masculino , Regeneración Nerviosa/fisiología , Ratas , Ratas Sprague-Dawley , Receptor para Productos Finales de Glicación Avanzada
7.
Neuropharmacology ; 65: 65-73, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23022047

RESUMEN

Diabetes causes mitochondrial dysfunction in sensory neurons that may contribute to peripheral neuropathy. Ciliary neurotrophic factor (CNTF) promotes sensory neuron survival and axon regeneration and prevents axonal dwindling, nerve conduction deficits and thermal hypoalgesia in diabetic rats. In this study, we tested the hypothesis that CNTF protects sensory neuron function during diabetes through normalization of impaired mitochondrial bioenergetics. In addition, we investigated whether the NF-κB signal transduction pathway was mobilized by CNTF. Neurite outgrowth of sensory neurons derived from streptozotocin (STZ)-induced diabetic rats was reduced compared to neurons from control rats and exposure to CNTF for 24 h enhanced neurite outgrowth. CNTF also activated NF-κB, as assessed by Western blotting for the NF-κB p50 subunit and reporter assays for NF-κB promoter activity. Conversely, blockade of NF-κB signaling using SN50 peptide inhibited CNTF-mediated neurite outgrowth. Studies in mice with STZ-induced diabetes demonstrated that systemic therapy with CNTF prevented functional indices of peripheral neuropathy along with deficiencies in dorsal root ganglion (DRG) NF-κB p50 expression and DNA binding activity. DRG neurons derived from STZ-diabetic mice also exhibited deficiencies in maximal oxygen consumption rate and associated spare respiratory capacity that were corrected by exposure to CNTF for 24 h in an NF-κB-dependent manner. We propose that the ability of CNTF to enhance axon regeneration and protect peripheral nerve from structural and functional indices of diabetic peripheral neuropathy is associated with targeting of mitochondrial function, in part via NF-κB activation, and improvement of cellular bioenergetics.


Asunto(s)
Factor Neurotrófico Ciliar/uso terapéutico , Diabetes Mellitus Experimental/metabolismo , Neuropatías Diabéticas/metabolismo , Metabolismo Energético/fisiología , FN-kappa B/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Células Cultivadas , Factor Neurotrófico Ciliar/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Neuropatías Diabéticas/prevención & control , Metabolismo Energético/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA