Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Aging Cell ; 20(1): e13287, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33369048

RESUMEN

Ischemic stroke, the third leading cause of death in the Western world, affects mainly the elderly and is strongly associated with comorbid conditions such as atherosclerosis or diabetes, which are pathologically characterized by increased inflammation and are known to influence the outcome of stroke. Stroke incidence peaks during influenza seasons, and patients suffering from infections such as pneumonia prior to stroke exhibit a worse stroke outcome. Earlier studies have shown that comorbidities aggravate the outcome of stroke, yet the mediators of this phenomenon remain obscure. Here, we show that acute peripheral inflammation aggravates stroke-induced neuronal damage and motor deficits specifically in aged mice. This is associated with increased levels of plasma proinflammatory cytokines, rather than with an increase of inflammatory mediators in the affected brain parenchyma. Nascent transcriptomics data with mature microRNA sequencing were used to identify the neuron-specific miRNome, in order to decipher dysregulated miRNAs in the brains of aged animals with stroke and co-existing inflammation. We pinpoint a previously uninvestigated miRNA in the brain, miR-127, that is highly neuronal, to be associated with increased cell death in the aged, LPS-injected ischemic mice. Target prediction tools indicate that miR-127 interacts with several basally expressed neuronal genes, and of these we verify miR-127 binding to Psmd3. Finally, we report reduced expression of miR-127 in human stroke brains. Our results underline the impact of peripheral inflammation on the outcome of stroke in aged subjects and pinpoint molecular targets for restoring endogenous neuronal capacity to combat ischemic stroke.


Asunto(s)
Isquemia Encefálica/genética , Inflamación/genética , MicroARNs/metabolismo , Envejecimiento , Animales , Isquemia Encefálica/mortalidad , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones
3.
J Neuroinflammation ; 17(1): 194, 2020 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-32560730

RESUMEN

BACKGROUND: Ischemic stroke is a devastating disease without a cure. The available treatments for ischemic stroke, thrombolysis by tissue plasminogen activator, and thrombectomy are suitable only to a fraction of patients and thus novel therapeutic approaches are urgently needed. The neuroinflammatory responses elicited secondary to the ischemic attack further aggravate the stroke-induced neuronal damage. It has been demonstrated that these responses are regulated at the level of non-coding RNAs, especially miRNAs. METHODS: We utilized lentiviral vectors to overexpress miR-669c in BV2 microglial cells in order to modulate their polarization. To detect whether the modulation of microglial activation by miR-669c provides protection in a mouse model of transient focal ischemic stroke, miR-669c overexpression was driven by a lentiviral vector injected into the striatum prior to induction of ischemic stroke. RESULTS: Here, we demonstrate that miR-669c-3p, a member of chromosome 2 miRNA cluster (C2MC), is induced upon hypoxic and excitotoxic conditions in vitro and in two different in vivo models of stroke. Rather than directly regulating the neuronal survival in vitro, miR-669c is capable of attenuating the microglial proinflammatory activation in vitro and inducing the expression of microglial alternative activation markers arginase 1 (Arg1), chitinase-like 3 (Ym1), and peroxisome proliferator-activated receptor gamma (PPAR-γ). Intracerebral overexpression of miR-669c significantly decreased the ischemia-induced cell death and ameliorated the stroke-induced neurological deficits both at 1 and 3 days post injury (dpi). Albeit miR-669c overexpression failed to alter the overall Iba1 protein immunoreactivity, it significantly elevated Arg1 levels in the ischemic brain and increased colocalization of Arg1 and Iba1. Moreover, miR-669c overexpression under cerebral ischemia influenced several morphological characteristics of Iba1 positive cells. We further demonstrate the myeloid differentiation primary response gene 88 (MyD88) transcript as a direct target for miR-669c-3p in vitro and show reduced levels of MyD88 in miR-669c overexpressing ischemic brains in vivo. CONCLUSIONS: Collectively, our data provide the evidence that miR-669c-3p is protective in a mouse model of ischemic stroke through enhancement of the alternative microglial/macrophage activation and inhibition of MyD88 signaling. Our results accentuate the importance of controlling miRNA-regulated responses for the therapeutic benefit in conditions of stroke and neuroinflammation.


Asunto(s)
Ventrículos Cerebrales/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , MicroARNs/metabolismo , Microglía/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Accidente Cerebrovascular Isquémico/genética , Ratones , MicroARNs/genética , Neuronas/metabolismo , Transducción de Señal/fisiología
4.
Neurochem Int ; 135: 104694, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32057948

RESUMEN

Research efforts in the past decades have provided insight into the adverse health effects of air pollution exposure. Exposure to airborne particulate matter is known to impair the respiratory and cardiovascular systems, and more recent investigations have provided evidence demonstrating harmful effects on the central nervous system. Investigations have primarily focused on the interconnected cellular pathways of inflammation and oxidative stress, which are induced by pollutant particle exposure both in peripheral tissues, and in the brain. Alterations to mitochondria, organelles important for cellular respiration and signaling, are often associated with increased cellular oxidative stress. This review focuses on the role of mitochondria in particulate matter-induced adverse effects on cellular health. More investigation to link air pollution and human health on the cellular and molecular level could in the future aid the development of more effective preventive and therapeutic options to combat pollutant particle-induced alterations.


Asunto(s)
Contaminación del Aire/efectos adversos , Encéfalo/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Material Particulado/efectos adversos , Animales , Encéfalo/metabolismo , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Material Particulado/metabolismo
5.
Neurotherapeutics ; 16(4): 1304-1319, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31372938

RESUMEN

Neuroinflammation is strongly induced by cerebral ischemia. The early phase after the onset of ischemic stroke is characterized by acute neuronal injury, microglial activation, and subsequent infiltration of blood-derived inflammatory cells, including macrophages. Therefore, modulation of the microglial/macrophage responses has increasingly gained interest as a potential therapeutic approach for the ischemic stroke. In our study, we investigated the effects of peripherally administered interleukin 13 (IL-13) in a mouse model of permanent middle cerebral artery occlusion (pMCAo). Systemic administration of IL-13 immediately after the ischemic insult significantly reduced the lesion volume, alleviated the infiltration of CD45+ leukocytes, and promoted the microglia/macrophage alternative activation within the ischemic region, as determined by arginase 1 (Arg1) immunoreactivity at 3 days post-ischemia (dpi). Moreover, IL-13 enhanced the expression of M2a alternative activation markers Arg1 and Ym1 in the peri-ischemic (PI) area, as well as increased plasma IL-6 and IL-10 levels at 3 dpi. Furthermore, IL-13 treatment ameliorated gait disturbances at day 7 and 14 and sensorimotor deficits at day 14 post-ischemia, as analyzed by the CatWalk gait analysis system and adhesive removal test, respectively. Finally, IL-13 treatment decreased neuronal cell death in a coculture model of neuroinflammation with RAW 264.7 macrophages. Taken together, delivery of IL-13 enhances microglial/macrophage anti-inflammatory responses in vivo and in vitro, decreases ischemia-induced brain cell death, and improves sensory and motor functions in the pMCAo mouse model of cerebral ischemia.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Interleucina-13/administración & dosificación , Macrófagos/efectos de los fármacos , Microglía/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Administración Intravenosa , Animales , Antiinflamatorios/administración & dosificación , Isquemia Encefálica/diagnóstico por imagen , Células Cultivadas , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Microglía/fisiología , Neuroprotección/fisiología , Accidente Cerebrovascular/diagnóstico por imagen
6.
Aging Dis ; 7(4): 450-65, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27493832

RESUMEN

Transient forebrain ischemia induces delayed death of the hippocampal pyramidal neurons, particularly in the CA2 and medial CA1 area. Early pharmacological inhibition of inflammatory response can ameliorate neuronal death, but it also inhibits processes leading to tissue regeneration. Therefore, research efforts are now directed to modulation of post-ischemic inflammation, with the aim to promote beneficial effects of inflammation and limit adverse effects. Transcription factor NF-κB plays a key role in the inflammation and cell survival/apoptosis pathways. In the brain, NF-κB is predominantly found in the form of a heterodimer of p65 (RelA) and p50 subunit, where p65 has a transactivation domain while p50 is chiefly involved in DNA binding. In this study, we subjected middle-aged Nfkb1 knockout mice (lacking p50 subunit) and wild-type controls of both sexs to 17 min of transient forebrain ischemia and assessed mouse performance in a panel of behavioral tests after two weeks of post-operative recovery. We found that ischemia failed to induce clear memory and motor deficits, but affected spontaneous locomotion in genotype- and sex-specific way. We also show that both the lack of the NF-κB p50 subunit and female sex independently protected CA2 hippocampal neurons from ischemia-induced cell death. Additionally, the NF-κB p50 subunit deficiency significantly reduced ischemia-induced microgliosis, astrogliosis, and neurogenesis. Lower levels of hippocampal microgliosis significantly correlated with faster spatial learning. We conclude that NF-κB regulates the outcome of transient forebrain ischemia in middle-aged subjects in a sex-specific way, having an impact not only on neuronal death but also specific inflammatory responses and neurogenesis.

7.
Glia ; 64(9): 1492-507, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27301579

RESUMEN

ADAMTS-4 (a disintegrin and metalloproteinase with thrombospondin motifs type 4) is a metalloprotease capable to degrade chondroitin sulfate proteoglycans leading to cartilage destruction during arthritis or to neuroplasticity during spinal cord injury (SCI). Although ADAMTS-4 is an inflammatory-regulated enzyme, its role during inflammation has never been investigated. The aim of this study was to investigate the role of ADAMTS-4 in neuroinflammation. First, we evidenced an increase of ADAMTS-4 expression in the ischemic brain hemisphere of mouse and human patients suffering from ischemic stroke. Then, we described that ADAMTS-4 has predominantly an anti-inflammatory effect in the CNS. Treatment of primary microglia or astrocyte cultures with low doses of a human recombinant ADAMTS-4 prior to LPS exposure decreased NO production and the synthesis/release of pro-inflammatory cytokines including NOS2, CCL2, TNF-α, IL-1ß and MMP-9. Accordingly, when cell cultures were transfected with silencing siRNA targeting ADAMTS-4 prior to LPS exposure, the production of NO and the synthesis/release of pro-inflammatory cytokines were increased. Finally, the feasibility of ADAMTS-4 to modulate neuroinflammation was investigated in vivo after permanent middle cerebral artery occlusion in mice. Although ADAMTS-4 treatment did not influence the lesion volume, it decreased astrogliosis and macrophage infiltration, and increased the number of microglia expressing arginase-1, a marker of alternatively activated cells with inflammation inhibiting functions. Additionally, ADAMTS-4 increased the production of IL-10 and IL-6 in the peri-ischemic area. By having anti-inflammatory and neuroregenerative roles, ADAMTS-4 may represent an interesting target to treat acute CNS injuries, such as ischemic stroke, SCI or traumatic brain injury. GLIA 2016;64:1492-1507.


Asunto(s)
Proteína ADAMTS4/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Modelos Animales de Enfermedad , Interleucina-10/metabolismo , Isquemia/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Microglía/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
8.
PLoS One ; 10(10): e0138583, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26426693

RESUMEN

Neuropathic pain treatment remains challenging due to ineffective therapy and resistance to opioid analgesia. Mitogen-activated protein kinase kinase (MAPKK) have been identified as the crucial regulators of pro- and antinociceptive factors. We used PD98059, an inhibitor of the MAPKK family members MEK1/2. The aim of study was to examine the influence of single and/or repeated PD98059 on nociception and opioid effectiveness in neuropathy. Moreover, we examined how PD98059 influences selected members of cellular pathways and cytokines. The PD98059 (2.5 mcg) was intrathecally preemptively administered before chronic constriction injury (CCI), and then once daily for 7 days. Additionally, at day 7 after CCI the PD98059-treated rats received a single injection of opioids. Using Western blot and qRT-PCR techniques in PD98059-treated rats we analyzed the mRNA and/or protein level of p38, ERK1/2, JNK, NF-kappaB, IL-1beta, IL-6, iNOS and IL-10 in the lumbar spinal cord. Our results indicate that PD98059 has an analgesic effects and potentiates morphine and/or buprenorphine analgesia. Parallel we observed that PD98059 inhibit upregulation of the CCI-elevated p38, ERK1/2, JNK and NF-kappaB protein levels. Moreover, PD98059 also prevented increase of pro- (IL-1beta, IL-6, and iNOS) but enhances anti-nociceptive (IL-10) factors. Summing up, PD98059 diminished pain and increased the effectiveness of opioids in neuropathy. The inhibition of MEKs might inactivate a variety of cell signaling pathways that are implicated in nociception.


Asunto(s)
Analgésicos Opioides/farmacología , Flavonoides/farmacología , Neuralgia/tratamiento farmacológico , Neuralgia/inmunología , Analgésicos Opioides/uso terapéutico , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , FN-kappa B/metabolismo , Neuralgia/fisiopatología , Nocicepción/efectos de los fármacos , Ratas , Ratas Wistar , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Neural Plast ; 2015: 676473, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26090236

RESUMEN

Neuropathic pain treatment remains a challenge because pathomechanism is not fully understood. It is believed that glial activation and increased spinal nociceptive factors are crucial for neuropathy. We investigated the effect of parthenolide (PTL) on the chronic constriction injury to the sciatic nerve (CCI)-induced neuropathy in rat. We analyzed spinal changes in glial markers and M1 and M2 polarization factors, as well as intracellular signaling pathways. PTL (5 µg; i.t.) was preemptively and then daily administered for 7 days after CCI. PTL attenuated the allodynia and hyperalgesia and increased the protein level of IBA1 (a microglial/macrophage marker) but did not change GFAP (an astrocyte marker) on day 7 after CCI. PTL reduced the protein level of M1 (IL-1ß, IL-18, and iNOS) and enhanced M2 (IL-10, TIMP1) factors. In addition, it downregulated the phosphorylated form of NF-κB, p38MAPK, and ERK1/2 protein level and upregulated STAT3. In primary microglial cell culture we have shown that IL-1ß, IL-18, iNOS, IL-6, IL-10, and TIMP1 are of microglial origin. Summing up, PTL directly or indirectly attenuates neuropathy symptoms and promotes M2 microglia/macrophages polarization. We suggest that neuropathic pain therapies should be shifted from blanketed microglia/macrophage suppression toward maintenance of the balance between neuroprotective and neurotoxic microglia/macrophage phenotypes.


Asunto(s)
Analgésicos/administración & dosificación , Macrófagos/efectos de los fármacos , Microglía/efectos de los fármacos , Neuralgia/metabolismo , Neuropatía Ciática/metabolismo , Sesquiterpenos/administración & dosificación , Analgésicos/uso terapéutico , Animales , Polaridad Celular/efectos de los fármacos , Células Cultivadas , Enfermedad Crónica , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Macrófagos/metabolismo , Masculino , Microglía/metabolismo , Neuralgia/tratamiento farmacológico , Ratas , Ratas Wistar , Neuropatía Ciática/tratamiento farmacológico , Sesquiterpenos/uso terapéutico , Transducción de Señal/efectos de los fármacos
10.
Mol Cell Neurosci ; 65: 1-10, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25662734

RESUMEN

Transient receptor potential vanilloid type 1 (TRPV1), classically associated with transduction of high-temperature and low-pH pain, underlies pain hypersensitivity in neuropathic pain. The molecular regulation of TRPV1 channel activity is not yet fully understood. Therefore, we investigated factors regulating sensitisation of this receptor during development of neuropathic pain in a rat model of chronic construction injury (CCI) in the dorsal root ganglia (DRG). In the rat CCI model, elevated levels of pro-inflammatory cytokines (TNFα, IL-1ß and IL-6) in DRG corresponded to development of neuropathic pain. We assessed the expression of known kinases influencing TRPV1 sensitisation at the mRNA and/or protein level. Protein kinase C ε (PKCε) showed the strongest upregulation at the mRNA and protein levels among all tested kinases. Co-expression of PKCε and TRPV1 in L5 DRG of CCI animals was high during the development of neuropathic pain. The number of neurons expressing PKCε increased throughout the experiment. We provide complex data on the expression of a variety of factors involved in TRPV1 sensitisation in a CCI model of neuropathic pain. Our study supports evidence for involvement of TRPV1 in the development of neuropathic pain, by showing increased expression of interleukins and kinases responsible for the channel sensitisation. TNFα and NGF seem to play a role in the transition from acute to neuropathic pain, while PKCε in its maintenance. Further studies might confirm their significance as novel targets for the treatment of neuropathic pain.


Asunto(s)
Neuralgia/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Ganglios Espinales/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Proteína Quinasa C-epsilon/genética , Proteína Quinasa C-epsilon/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Canales Catiónicos TRPV/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
11.
Pain ; 156(5): 890-903, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25719612

RESUMEN

The pharmacological inhibition of anandamide (AEA) hydrolysis by fatty acid amide hydrolase (FAAH) attenuates pain in animal models of osteoarthritis (OA) but has failed in clinical trials. This may have occurred because AEA also activates transient receptor potential vanilloid type 1 (TRPV1), which contributes to pain development. Therefore, we investigated the effectiveness of the dual FAAH-TRPV1 blocker OMDM-198 in an MIA-model of osteoarthritic pain. We first investigated the MIA-induced model of OA by (1) characterizing the pain phenotype and degenerative changes within the joint using X-ray microtomography and (2) evaluating nerve injury and inflammation marker (ATF-3 and IL-6) expression in the lumbar dorsal root ganglia of osteoarthritic rats and differences in gene and protein expression of the cannabinoid CB1 receptors FAAH and TRPV1. Furthermore, we compared OMDM-198 with compounds acting exclusively on FAAH or TRPV1. Osteoarthritis was accompanied by the fragmentation of bone microstructure and destroyed cartilage. An increase of the mRNA levels of ATF3 and IL-6 and an upregulation of AEA receptors and FAAH in the dorsal root ganglia were observed. OMDM-198 showed antihyperalgesic effects in the OA model, which were comparable with those of a selective TRPV1 antagonist, SB-366,791, and a selective FAAH inhibitor, URB-597. The effect of OMDM-198 was attenuated by the CB1 receptor antagonist, AM-251, and by the nonpungent TRPV1 agonist, olvanil, suggesting its action as an "indirect" CB1 agonist and TRPV1 antagonist. These results suggest an innovative strategy for the treatment of OA, which may yield more satisfactory results than those obtained so far with selective FAAH inhibitors in human OA.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Carbamatos/administración & dosificación , Carbamatos/farmacología , Ganglios Espinales/metabolismo , Osteoartritis/fisiopatología , Dolor/tratamiento farmacológico , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/metabolismo , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Anilidas/administración & dosificación , Anilidas/farmacología , Animales , Ácidos Araquidónicos/metabolismo , Benzamidas/farmacología , Capsaicina/análogos & derivados , Capsaicina/farmacología , Cinamatos/administración & dosificación , Cinamatos/farmacología , Modelos Animales de Enfermedad , Endocannabinoides/metabolismo , Ganglios Espinales/citología , Expresión Génica/efectos de los fármacos , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/genética , Hiperalgesia/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Interleucina-6/genética , Interleucina-6/metabolismo , Vértebras Lumbares/citología , Masculino , Dolor/inmunología , Dolor/metabolismo , Manejo del Dolor/métodos , Dimensión del Dolor/métodos , Piperidinas/farmacología , Alcamidas Poliinsaturadas/metabolismo , Pirazoles/farmacología , Ratas , Ratas Wistar , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/metabolismo , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA