Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
PLoS Genet ; 18(2): e1009644, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35139074

RESUMEN

Transcription of the ~200 mouse and human ribosomal RNA genes (rDNA) by RNA Polymerase I (RPI/PolR1) accounts for 80% of total cellular RNA, around 35% of all nuclear RNA synthesis, and determines the cytoplasmic ribosome complement. It is therefore a major factor controlling cell growth and its misfunction has been implicated in hypertrophic and developmental disorders. Activation of each rDNA repeat requires nucleosome replacement by the architectural multi-HMGbox factor UBTF to create a 15.7 kbp nucleosome free region (NFR). Formation of this NFR is also essential for recruitment of the TBP-TAFI factor SL1 and for preinitiation complex (PIC) formation at the gene and enhancer-associated promoters of the rDNA. However, these promoters show little sequence commonality and neither UBTF nor SL1 display significant DNA sequence binding specificity, making what drives PIC formation a mystery. Here we show that cooperation between SL1 and the longer UBTF1 splice variant generates the specificity required for rDNA promoter recognition in cell. We find that conditional deletion of the TAF1B subunit of SL1 causes a striking depletion of UBTF at both rDNA promoters but not elsewhere across the rDNA. We also find that while both UBTF1 and -2 variants bind throughout the rDNA NFR, only UBTF1 is present with SL1 at the promoters. The data strongly suggest an induced-fit model of RPI promoter recognition in which UBTF1 plays an architectural role. Interestingly, a recurrent UBTF-E210K mutation and the cause of a pediatric neurodegeneration syndrome provides indirect support for this model. E210K knock-in cells show enhanced levels of the UBTF1 splice variant and a concomitant increase in active rDNA copies. In contrast, they also display reduced rDNA transcription and promoter recruitment of SL1. We suggest the underlying cause of the UBTF-E210K syndrome is therefore a reduction in cooperative UBTF1-SL1 promoter recruitment that may be partially compensated by enhanced rDNA activation.


Asunto(s)
Proteínas del Complejo de Iniciación de Transcripción Pol1 , ARN Polimerasa I , Animales , Niño , ADN Ribosómico/genética , ADN Ribosómico/metabolismo , Humanos , Ratones , Nucleosomas , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , Regiones Promotoras Genéticas , ARN Polimerasa I/genética , ARN Ribosómico/genética , Transcripción Genética
2.
Int J Mol Sci ; 25(4)2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38396946

RESUMEN

Neurodegenerative diseases (NDDs), including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), are gradually becoming a burden to society. The adverse effects and mortality/morbidity rates associated with these NDDs are a cause of many healthcare concerns. The pathologic alterations of NDDs are related to mitochondrial dysfunction, oxidative stress, and inflammation, which further stimulate the progression of NDDs. Recently, long non-coding RNAs (lncRNAs) have attracted ample attention as critical mediators in the pathology of NDDs. However, there is a significant gap in understanding the biological function, molecular mechanisms, and potential importance of lncRNAs in NDDs. This review documents the current research on lncRNAs and their implications in NDDs. We further summarize the potential implication of lncRNAs to serve as novel therapeutic targets and biomarkers for patients with NDDs.


Asunto(s)
Enfermedad de Alzheimer , Esclerosis Amiotrófica Lateral , Enfermedades Neurodegenerativas , Enfermedad de Parkinson , ARN Largo no Codificante , Humanos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , ARN Largo no Codificante/genética , Enfermedad de Parkinson/genética , Esclerosis Amiotrófica Lateral/genética
3.
Neurobiol Dis ; 156: 105399, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34029695

RESUMEN

Immune system hypersensitivity is believed to contribute to mental frailty in the elderly. Solid evidence indicates NOD-like receptor pyrin domain containing-3 (NLRP3)-inflammasome activation intimately connects aging-associated chronic inflammation (inflammaging) to senile cognitive decline. Thioredoxin interacting protein (TXNIP), an inducible protein involved in oxidative stress, is essential for NLRP3 inflammasome activity. This study aims to find whether TXNIP/NLRP3 inflammasome pathway is involved in senile dementia. According to our studies on sex-matched mice, TXNIP was significantly upregulated in aged animals, paralleled by the NLRP3-inflammasome over-activity leading to enhanced caspase-1 cleavage and IL-1ß maturation, in both sexes. This was closely associated with depletion of the anti-aging and cognition enhancing protein klotho, in aged males. Txnip knockout reversed age-related NLRP3-hyperactivity and enhanced thioredoxin (TRX) levels. Further, TXNIP inhibition along with verapamil replicated TXNIP/NLRP3-inflammasome downregulation in aged animals, with FOXO-1 and mTOR upregulation. These alterations concurred with substantial improvements in both cognitive and sensorimotor abilities. Together, these findings substantiate the pivotal role of TXNIP to drive inflammaging in parallel with klotho depletion and functional decline, and delineate thioredoxin system as a potential target to decelerate senile dementia.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Proteínas Portadoras/biosíntesis , Mediadores de Inflamación/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/biosíntesis , Tiorredoxinas/biosíntesis , Envejecimiento/genética , Envejecimiento/patología , Animales , Encéfalo/patología , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Femenino , Mediadores de Inflamación/antagonistas & inhibidores , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Estrés Oxidativo/fisiología , Tiorredoxinas/antagonistas & inhibidores , Tiorredoxinas/genética
4.
Chromosome Res ; 27(4): 345-364, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31707536

RESUMEN

The complexity of neurodegeneration restricts the ability to understand and treat the neurological disorders affecting millions of people worldwide. Therefore, there is an unmet need to develop new and more effective therapeutic strategies to combat these devastating conditions and that will only be achieved with a better understanding of the biological mechanism associated with disease conditions. Recent studies highlight the role of DNA damage, particularly, DNA double-strand breaks (DSBs), in the progression of neuronal loss in a broad spectrum of human neurodegenerative diseases. This is not unexpected because neurons are prone to DNA damage due to their non-proliferative nature and high metabolic activity. However, it is not clear if DSBs is a primary driver of neuronal loss in disease conditions or simply occurs concomitant with disease progression. Here, we provide evidence that supports a critical role of DSBs in the pathogenesis of the neurodegenerative diseases. Among different kinds of DNA damages, DSBs are the most harmful and perilous type of DNA damage and can lead to cell death if left unrepaired or repaired with error. In this review, we explore the current state of knowledge regarding the role of DSBs repair mechanisms in preserving neuronal function and survival and describe how DSBs could drive the molecular mechanisms resulting in neuronal death in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. We also discuss the potential implications of DSBs as a novel therapeutic target and prognostic marker in patients with neurodegenerative conditions.


Asunto(s)
Roturas del ADN de Doble Cadena , Predisposición Genética a la Enfermedad , Enfermedades Neurodegenerativas/genética , Animales , Biomarcadores , Daño del ADN , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Diagnóstico Diferencial , Estudios de Asociación Genética , Pruebas Genéticas , Recombinación Homóloga , Humanos , Terapia Molecular Dirigida , Enfermedades Neurodegenerativas/diagnóstico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/terapia
5.
Mol Med ; 21: 637-44, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-26252187

RESUMEN

Sepsis and septic shock are enormous public health problems with astronomical financial repercussions on health systems worldwide. The central nervous system (CNS) is closely intertwined in the septic process but the underlying mechanism is still obscure. AMP-activated protein kinase (AMPK) is a ubiquitous energy sensor enzyme and plays a key role in regulation of energy homeostasis and cell survival. In this study, we hypothesized that activation of AMPK in the brain would attenuate inflammatory responses in sepsis, particularly in the lungs. Adult C57BL/6 male mice were treated with 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR, 20 ng), an AMPK activator, or vehicle (normal saline) by intracerebroventricular (ICV) injection, followed by cecal ligation and puncture (CLP) at 30 min post-ICV. The septic mice treated with AICAR exhibited elevated phosphorylation of AMPKα in the brain along with reduced serum levels of aspartate aminotransferase, tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and interleukin-6 (IL-6), compared with the vehicle. Similarly, the expressions of TNF-α, IL-1ß, keratinocyte-derived chemokine and macrophage inflammatory protein-2 as well as myeloperoxidase activity in the lungs of AICAR-treated mice were significantly reduced. Moreover, histological findings in the lungs showed improvement of morphologic features and reduction of apoptosis with AICAR treatment. We further found that the beneficial effects of AICAR on septic mice were diminished in AMPKα2 deficient mice, showing that AMPK mediates these effects. In conclusion, our findings reveal a new functional role of activating AMPK in the CNS to attenuate inflammatory responses and acute lung injury in sepsis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/biosíntesis , Lesión Pulmonar Aguda/genética , Inflamación/genética , Sepsis/genética , Proteínas Quinasas Activadas por AMP/genética , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/terapia , Aminoimidazol Carboxamida/administración & dosificación , Aminoimidazol Carboxamida/análogos & derivados , Animales , Encéfalo/metabolismo , Supervivencia Celular/efectos de los fármacos , Metabolismo Energético/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/patología , Inflamación/terapia , Masculino , Ratones , Fosforilación/efectos de los fármacos , Ribonucleótidos/administración & dosificación , Sepsis/metabolismo , Sepsis/patología , Sepsis/terapia
6.
Neurochem Res ; 40(5): 980-90, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25754447

RESUMEN

Previously, we have shown that aberrant expression of glia maturation factor (GMF), a proinflammatory protein, is associated with the neuropathological conditions underlying diseases suggesting an important role for GMF in neurodegeneration. In the present study, we demonstrate that absence of GMF suppresses dopaminergic (DA) neuron loss, glial activation, and expression of proinflammatory mediators in the substantia nigra pars compacta (SN) and striatum (STR) of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treated mice. Dopaminergic neuron numbers in the SN and fiber densities in the STR were reduced in wild type (Wt) mice when compared with GMF-deficient (GMF-KO) mice after MPTP treatment. We compared the motor abnormalities caused by MPTP treatment in Wt and GMF-KO mice as measured by Rota rod and grip strength test. Results show that the deficits in motor coordination and decrease in dopamine and its metabolite content were protected significantly in GMF-KO mice after MPTP treatment when compared with control Wt mice under identical experimental conditions. These findings were further supported by the immunohistochemical analysis that showed reduced glial activation in the SN of MPTP-treated GMF-KO mice. Similarly, in MPTP-treated GMF-KO mice, production of inflammatory tumor necrosis factor alpha, interleukine-1 beta, granulocyte macrophage-colony stimulating factor, and the chemokine (C-C motif) ligand 2 MCP-1 was suppressed, findings consistent with a role for GMF in MPTP neurotoxicity. In conclusion, present investigation provides the first evidence that deficiency of GMF protects the DA neuron loss and reduces the inflammatory load following MPTP administration in mice. Thus depletion of endogenous GMF represents an effective and selective strategy to slow down the MPTP-induced neurodegeneration.


Asunto(s)
Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Factor de Maduración de la Glia/deficiencia , Actividad Motora/fisiología , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/prevención & control , Animales , Neuronas Dopaminérgicas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trastornos Parkinsonianos/patología
7.
Blood ; 119(10): 2385-91, 2012 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-22123843

RESUMEN

ADAMTS13, a metalloprotease, plays a pivotal role in preventing spontaneous microvascular thrombosis by cleaving hyperactive ultra large von Willebrand factor multimers into smaller, less active multimers. Reduced ADAMTS13 activity in plasma has been described in many diseases associated with systemic inflammation. It remains uncertain, however, whether ADAMTS13 contributes to disease pathogenesis or rather simply serves as an inflammation-associated marker. We hypothesized that, by decreasing vascular inflammation, ADAMTS13 reduces the development of early atherosclerotic plaques. Using intravital fluorescence microscopy, we observed excessive leukocyte adhesion and accelerated atherosclerotic plaque formation at the carotid sinus of Adamts13(-/-)/ApoE(-/-) mice compared with ApoE(-/-) mice fed a high-fat Western diet. At 4 months of age, there was a significant increase in atherosclerosis in the aorta and aortic sinus of Adamts13(-/-)/ApoE(-/-) mice compared with ApoE(-/-) mice. Interestingly, we detected a 2-fold increase in macrophage recruitment to the atherosclerotic plaque of the Adamts13(-/-)/ApoE(-/-) mice compared with ApoE(-/-) mice, suggesting that the atherosclerotic lesions in these mice were not only larger but also more inflammatory. These findings reveal a new functional role for the antithrombotic enzyme ADAMTS13 in reducing excessive vascular inflammation and plaque formation during early atherosclerosis.


Asunto(s)
Apolipoproteínas E/metabolismo , Metaloendopeptidasas/metabolismo , Placa Aterosclerótica/metabolismo , Vasculitis/metabolismo , Proteína ADAMTS13 , Animales , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/genética , Adhesión Celular , Colágeno Tipo I/metabolismo , Colágeno Tipo II/metabolismo , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/toxicidad , Femenino , Inmunohistoquímica , Leucocitos/metabolismo , Leucocitos/patología , Lípidos/sangre , Macrófagos/metabolismo , Masculino , Metaloendopeptidasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Placa Aterosclerótica/etiología , Placa Aterosclerótica/genética , Seno Aórtico/metabolismo , Seno Aórtico/patología , Factores de Tiempo , Vasculitis/sangre , Vasculitis/genética
8.
Neural Regen Res ; 19(5): 1020-1026, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-37862204

RESUMEN

Tauopathies are a group of neurological disorders, including Alzheimer's disease and frontotemporal dementia, which involve progressive neurodegeneration, cognitive deficits, and aberrant tau protein accumulation. The development of tauopathies cannot currently be stopped or slowed down by treatment measures. Given the significant contribution of tau burden in primary tauopathies and the strong association between pathogenic tau accumulation and cognitive deficits, there has been a lot of interest in creating therapies that can alleviate tau pathology and render neuroprotective effects. Recently, small molecules, immunotherapies, and gene therapy have been used to reduce the pathological tau burden and prevent neurodegeneration in animal models of tauopathies. However, the major pitfall of the current therapeutic approach is the difficulty of drugs and gene-targeting modalities to cross the blood-brain barrier and their unintended side effects. In this review, the current therapeutic strategies used for tauopathies including the use of oligonucleotide-based gene therapy approaches that have shown a promising result for the treatment of tauopathies and Alzheimer's disease in preclinical animal models, have been discussed.

9.
Brain Sci ; 14(2)2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38391753

RESUMEN

Recently, a recurrent de novo dominant mutation in UBTF (c.628G>A, p.Glu210Lys; UBTF E210K) was identified as the cause of a neurological disorder which has been named UBTF Neuroregression Syndrome (UNS), or Childhood-Onset Neurodegeneration with Brain Atrophy (CONDBA). To date, only 17 cases have been reported worldwide. The molecular etiology is a pathogenic variant, E210K, within the HMG-box 2 of Upstream Binding Transcription Factor (UBTF). UBTF, a nucleolar protein, plays an important role in ribosomal RNA (rRNA) synthesis, nucleolar integrity, and cell survival. This variant causes unstable preinitiation complexes to form, resulting in altered rDNA chromatin structures, rRNA dysregulation, DNA damage, and ultimately, neurodegeneration. Defining clinical characteristics of the disorder include but are not limited to developmental regression beginning at approximately three years of age, progressive motor dysfunction, declining cognition, ambulatory loss, and behavioral problems. Histological and neuroimaging abnormalities include cortical atrophy, white matter deficits, and enlarged ventricles. Herein, we present a detailed overview of all published cases as well as the functional roles of UBTF to better understand the pathophysiology. Bringing undiagnosed cases to the attention of clinicians and researchers by making them aware of the clinical features will improve research and support the development of therapeutic interventions.

10.
Neurochem Res ; 38(1): 218-25, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23086473

RESUMEN

We previously demonstrated that glia maturation factor (GMF), a brain specific protein, isolated, sequenced and cloned in our laboratory, induce expression of proinflammatory cytokines and chemokines in the central nervous system. We also reported that the up-regulation of GMF in astrocytes leads to the destruction of neurons suggesting a novel pathway of GMF-mediated cytotoxicity of brain cells, and implicated its involvement in the pathogenesis of inflammatory neurodegenerative diseases. In the present study, we examined the expressions of GMF in triple-transgenic Alzheimer's disease (3xTg-AD) mice. Our results show a 13-fold up-regulation of GMF and 8-12-fold up-regulation of proinflammatory cytokines tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), IL-1ß, interferon gamma (IFN-γ), and chemokine (C-C motif) ligand 2 (CCL2) and C-X-C motif chemokine 10 (CXCL10/IP-10) mRNA as determined by quantitative real-time RT-PCR in the brain of 3xTg-AD mice as compared to non-transgenic (Non-Tg) mice. In conclusion, the increase in GMF and cytokine/chemokine expression was correlated with reactive glial fibrillary acidic protein positive astrocytes and ionized calcium binding adaptor molecule 1 (Iba-1)-positive microglia in 3xTg-AD mice.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Encéfalo/inmunología , Factor de Maduración de la Glia/biosíntesis , Activación de Macrófagos/genética , Neuroglía/fisiología , Enfermedad de Alzheimer/psicología , Precursor de Proteína beta-Amiloide/genética , Animales , Western Blotting , Encefalopatías/genética , Encefalopatías/patología , Quimiocinas/biosíntesis , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/psicología , Citocinas/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Potenciación a Largo Plazo/genética , Potenciación a Largo Plazo/fisiología , Ratones , Ratones Transgénicos , Ovillos Neurofibrilares/genética , Ovillos Neurofibrilares/patología , Neuroglía/inmunología , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Placa Amiloide/genética , Placa Amiloide/patología , Presenilina-1/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas tau/genética
11.
Curr Neuropharmacol ; 21(5): 1100-1116, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36089791

RESUMEN

Neurodegenerative and neurovascular disorders affect millions of people worldwide and account for a large and increasing health burden on the general population. Thus, there is a critical need to identify potential disease-modifying treatments that can prevent or slow the disease progression. Mitochondria are highly dynamic organelles and play an important role in energy metabolism and redox homeostasis, and mitochondrial dysfunction threatens cell homeostasis, perturbs energy production, and ultimately leads to cell death and diseases. Impaired mitochondrial function has been linked to the pathogenesis of several human neurological disorders. Given the significant contribution of mitochondrial dysfunction in neurological disorders, there has been considerable interest in developing therapies that can attenuate mitochondrial abnormalities and proffer neuroprotective effects. Unfortunately, therapies that target specific components of mitochondria or oxidative stress pathways have exhibited limited translatability. To this end, mitochondrial transplantation therapy (MTT) presents a new paradigm of therapeutic intervention, which involves the supplementation of healthy mitochondria to replace the damaged mitochondria for the treatment of neurological disorders. Prior studies demonstrated that the supplementation of healthy donor mitochondria to damaged neurons promotes neuronal viability, activity, and neurite growth and has been shown to provide benefits for neural and extra-neural diseases. In this review, we discuss the significance of mitochondria and summarize an overview of the recent advances and development of MTT in neurodegenerative and neurovascular disorders, particularly Parkinson's disease, Alzheimer's disease, and stroke. The significance of MTT is emerging as they meet a critical need to develop a diseasemodifying intervention for neurodegenerative and neurovascular disorders.


Asunto(s)
Mitocondrias , Enfermedades Neurodegenerativas , Humanos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/terapia , Metabolismo Energético , Mitocondrias/metabolismo , Mitocondrias/patología , Mitocondrias/trasplante , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/terapia , Neuronas/metabolismo , Neuronas/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/terapia , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/terapia , Animales
12.
J Cachexia Sarcopenia Muscle ; 14(1): 493-507, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36604839

RESUMEN

BACKGROUND: Injection of exogenous mitochondria has been shown to improve the ischaemia-damaged myocardium, but the effect of mitochondrial transplant therapy (MTT) to restore skeletal muscle mass and function has not been tested following neuromuscular injury. Therefore, we tested the hypothesis that MTT would improve the restoration of muscle function after injury. METHODS: BaCl2 was injected into the gastrocnemius muscle of one limb of 8-12-week-old C57BL/6 mice to induce damage without injury to the resident stem cells. The contralateral gastrocnemius muscle was injected with phosphate-buffered saline (PBS) and served as the non-injured intra-animal control. Mitochondria were isolated from donor mice. Donor mitochondria were suspended in PBS or PBS without mitochondria (sham treatment) and injected into the tail vein of BaCl2 injured mice 24 h after the initial injury. Muscle repair was examined 7, 14 and 21 days after injury. RESULTS: MTT did not increase systemic inflammation in mice. Muscle mass 7 days following injury was 21.9 ± 2.1% and 17.4 ± 1.9% lower (P < 0.05) in injured as compared with non-injured intra-animal control muscles in phosphate-buffered saline (PBS)- and MTT-treated animals, respectively. Maximal plantar flexor muscle force was significantly lower in injured as compared with uninjured muscles of PBS-treated (-43.4 ± 4.2%, P < 0.05) and MTT-treated mice (-47.7 ± 7.3%, P < 0.05), but the reduction in force was not different between the experimental groups. The percentage of collagen and other non-contractile tissue in histological muscle cross sections, was significantly greater in injured muscles of PBS-treated mice (33.2 ± 0.2%) compared with MTT-treated mice (26.5 ± 0.2%) 7 days after injury. Muscle wet weight and maximal muscle force from injured MTT-treated mice had recovered to control levels by 14 days after the injury. However, muscle mass and force had not improved in PBS-treated animals by 14 days after injury. The non-contractile composition of the gastrocnemius muscle tissue cross sections was not different between control, repaired PBS-treated and repaired MTT-treated mice 14 days after injury. By 21 days following injury, PBS-treated mice had fully restored gastrocnemius muscle mass of the injured muscle to that of the uninjured muscle, although maximal plantar flexion force was still 19.4 ± 3.7% (P < 0.05) lower in injured/repaired gastrocnemius as compared with uninjured intra-animal control muscles. CONCLUSIONS: Our results suggest that systemic mitochondria delivery can enhance the rate of muscle regeneration and restoration of muscle function following injury.


Asunto(s)
Enfermedades Musculares , Regeneración , Ratones , Animales , Ratones Endogámicos C57BL , Músculo Esquelético/patología , Enfermedades Musculares/metabolismo , Mitocondrias , Fosfatos/metabolismo , Fosfatos/farmacología
13.
Stroke ; 43(5): 1376-82, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22363055

RESUMEN

BACKGROUND AND PURPOSE: The fibronectin isoform containing the alternatively spliced extra domain A (EDA(+)-FN) is normally absent from the circulation, but plasma levels of EDA(+)-FN can become markedly elevated in several human pathological conditions associated with inflammation including ischemic stroke. It remains unknown whether EDA(+)-FN contributes to stroke pathogenesis or is simply an associative marker. Several in vitro studies suggest that EDA(+)-FN can activate Toll-like receptor 4, an innate immune receptor that triggers proinflammatory responses. We undertook a genetic approach in mice to investigate the ability of EDA(+)-FN to mediate inflammatory brain damage in a focal cerebral ischemia/reperfusion injury model. METHODS: We used genetically modified EDA(+/+) mice, which constitutively express EDA(+)-FN. Extent of injury, neurological outcome, and inflammatory mechanisms were assessed after 1-hour cerebral ischemia/23-hour reperfusion injury and compared with wild-type mice. RESULTS: We found that EDA(+/+) mice developed significantly larger infarcts and severe neurological deficits that were associated with significant increased neutrophil and macrophage infiltration as quantitated by immunohistochemistry. Additionally, we found upregulation of nuclear factor-κB, cyclo-oxygenase-2, and inflammatory cytokines tumor necrosis factor-α, interleukin-1ß, and interleukin-6 in the EDA(+/+) mice compared with wild-type mice. Interestingly, increased brain injury and neurological deficits were largely abrogated in EDA(+/+) mice by treatment with a specific Toll-like receptor 4 inhibitor. CONCLUSIONS: These findings provide the first evidence that EDA(+)-FN promotes inflammatory brain injury after ischemic stroke and suggest that the elevated levels of plasma EDA(+)-FN observed in chronic inflammatory conditions could worsen injury and outcome in patients after acute stroke.


Asunto(s)
Empalme Alternativo/genética , Lesiones Encefálicas/fisiopatología , Isquemia Encefálica/fisiopatología , Encefalitis/fisiopatología , Fibronectinas/genética , Fibronectinas/fisiología , Estructura Terciaria de Proteína/genética , Animales , Lesiones Encefálicas/sangre , Lesiones Encefálicas/etiología , Isquemia Encefálica/sangre , Isquemia Encefálica/complicaciones , Ciclooxigenasa 2/fisiología , Citocinas/fisiología , Encefalitis/sangre , Encefalitis/etiología , Fibronectinas/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , FN-kappa B/fisiología , Factores de Tiempo , Receptor Toll-Like 4/fisiología , Regulación hacia Arriba/fisiología
14.
Mol Neurobiol ; 58(1): 118-131, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32895786

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease that accounts for a majority of dementia cases. AD is characterized by progressive neuronal death associated with neuropathological lesions consisting of neurofibrillary tangles and senile plaques. While the pathogenesis of AD has been widely investigated, significant gaps in our knowledge remain about the cellular and molecular mechanisms promoting AD. Recent studies have highlighted the role of DNA damage, particularly DNA double-strand breaks (DSBs), in the progression of neuronal loss in a broad spectrum of neurodegenerative diseases. In the present study, we tested the hypothesis that accumulation of DNA DSB plays an important role in AD pathogenesis. To test our hypothesis, we examined DNA DSB expression and DNA repair function in the hippocampus of human AD and non-AD brains by immunohistochemistry, ELISA, and RT-qPCR. We observed increased DNA DSB accumulation and reduced DNA repair function in the hippocampus of AD brains compared to the non-AD control brains. Next, we found significantly increased levels of DNA DSB and altered levels of DNA repair proteins in the hippocampus of 5xFAD mice compared to non-transgenic mice. Interestingly, increased accumulation of DNA DSBs and altered DNA repair proteins were also observed in cellular models of AD. These findings provided compelling evidence that AD is associated with accumulation of DNA DSB and/or alteration in DSB repair proteins which may influence an important early part of the pathway toward neural damage and memory loss in AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/patología , Roturas del ADN de Doble Cadena , Cambios Post Mortem , Animales , Células CHO , Células Cultivadas , Cricetulus , Reparación del ADN , Modelos Animales de Enfermedad , Hipocampo/patología , Humanos , Ratones Transgénicos
15.
J Neuroimmune Pharmacol ; 16(3): 667-678, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33221984

RESUMEN

Parkinson's disease (PD) is characterized by progressive degeneration of dopaminergic neurons in the substantia nigra and loss of both motor and non-motor features. Several clinical and preclinical studies have provided evidence that estrogen therapy reduces the risk of PD but have limitations in terms of adverse peripheral effects. Therefore, we examined the potential beneficial effects of the brain-selective estrogen prodrug, 10ß, 17ß-dihydroxyestra-1,4-dien-3-one (DHED) on nigrostriatal dopaminergic neurodegeneration and behavioral abnormalities in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. Wild-type mice were treated with daily subcutaneous injections of DHED (50 and 100 µg/kg) or vehicle for four weeks. To produce PD-like symptoms, mice were injected with MPTP (18 mg/kg in saline; intraperitoneally) four times at 2-hr intervals for one day. After behavioral examination, mice were sacrificed, and the brains were isolated for neurochemical and morphological examinations. MPTP injected mice exhibited loss of dopaminergic neurons and fibers in substantia nigra and striatum respectively, along with impaired motor function at day 7 post MPTP injection. These phenotypes were associated with significantly increased oxidative stress and inflammatory responses in the striatum regions. DHED treatments significantly mitigated behavioral impairments and dopaminergic neurodegeneration induced by MPTP. We further observed that DHED treatment suppressed oxidative stress and inflammation in the striatum of MPTP treated mice when compared to vehicle treated mice. In conclusions, our findings suggest that DHED protects dopaminergic neurons from MPTP toxicity in mouse model of PD and support a beneficial effect of brain-selective estrogen in attenuating neurodegeneration and motor symptoms in PD-related neurological disorders. Graphical Abstract.


Asunto(s)
Intoxicación por MPTP , Enfermedad de Parkinson , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , Encéfalo , Cuerpo Estriado , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas , Estrógenos/farmacología , Intoxicación por MPTP/tratamiento farmacológico , Intoxicación por MPTP/prevención & control , Ratones , Ratones Endogámicos C57BL , Sustancia Negra
16.
Cells ; 10(4)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33916001

RESUMEN

Alzheimer's disease (AD), a progressive neurodegenerative disorder characterized by memory loss and cognitive decline, is a major cause of death and disability among the older population. Despite decades of scientific research, the underlying etiological triggers are unknown. Recent studies suggested that gut microbiota can influence AD progression; however, potential mechanisms linking the gut microbiota with AD pathogenesis remain obscure. In the present study, we provided a potential mechanistic link between dysbiotic gut microbiota and neuroinflammation associated with AD progression. Using a mouse model of AD, we discovered that unfavorable gut microbiota are correlated with abnormally elevated expression of gut NLRP3 and lead to peripheral inflammasome activation, which in turn exacerbates AD-associated neuroinflammation. To this end, we observe significantly altered gut microbiota compositions in young and old 5xFAD mice compared to age-matched non-transgenic mice. Moreover, 5xFAD mice demonstrated compromised gut barrier function as evident from the loss of tight junction and adherens junction proteins compared to non-transgenic mice. Concurrently, we observed increased expression of NLRP3 inflammasome and IL-1ß production in the 5xFAD gut. Consistent with our hypothesis, increased gut-microbial-inflammasome activation is positively correlated with enhanced astrogliosis and microglial activation, along with higher expression of NLRP3 inflammasome and IL-1ß production in the brains of 5xFAD mice. These data indicate that the elevated expression of gut-microbial-inflammasome components may be an important trigger for subsequent downstream activation of inflammatory and potentially cytotoxic mediators, and gastrointestinal NLRP3 may promote NLRP3 inflammasome-mediated neuroinflammation. Thus, modulation of the gut microbiota may be a potential strategy for the treatment of AD-related neurological disorders in genetically susceptible hosts.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/microbiología , Encéfalo/metabolismo , Microbioma Gastrointestinal , Inflamasomas/metabolismo , Envejecimiento/patología , Enfermedad de Alzheimer/patología , Animales , Apoptosis , Encéfalo/patología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasa 1/metabolismo , Modelos Animales de Enfermedad , Tracto Gastrointestinal/patología , Inflamación/patología , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Unión a Fosfato/metabolismo
17.
Pharmaceutics ; 13(4)2021 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-33917577

RESUMEN

The blood-brain barrier (BBB) is a natural obstacle for drug delivery into the human brain, hindering treatment of central nervous system (CNS) disorders such as acute ischemic stroke, brain tumors, and human immunodeficiency virus (HIV)-1-associated neurocognitive disorders. Poly(lactic-co-glycolic acid) (PLGA) is a biocompatible polymer that is used in Food and Drug Administration (FDA)-approved pharmaceutical products and medical devices. PLGA nanoparticles (NPs) have been reported to improve drug penetration across the BBB both in vitro and in vivo. Poly(ethylene glycol) (PEG), poly(vinyl alcohol) (PVA), and poloxamer (Pluronic) are widely used as excipients to further improve the stability and effectiveness of PLGA formulations. Peptides and other linkers can be attached on the surface of PLGA to provide targeting delivery. With the newly published guidance from the FDA and the progress of current Good Manufacturing Practice (cGMP) technologies, manufacturing PLGA NP-based drug products can be achieved with higher efficiency, larger quantity, and better quality. The translation from bench to bed is feasible with proper research, concurrent development, quality control, and regulatory assurance.

18.
Behav Pharmacol ; 21(5-6): 563-71, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20657266

RESUMEN

Increased oxidative stress is implicated in the pathogenesis of Parkinson's disease in which dopaminergic neurons are intrinsically susceptible to oxidative damage. Swiss albino mice were pretreated with Pycnogenol (PYC), an extract of Pinus maritime bark [20 mg/kg body weight, intraperitoneally (i.p.)] once daily for 15 days. Thereafter, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) (20 mg/kg body weight, intraperitoneally) was given four times at 2-hour intervals on 1 day only. Behaviours were altered in the MPTP group as compared with the vehicle-treated group and were restored in the PYC-pretreated MPTP group. The activity of antioxidant enzymes and the content of glutathione were significantly depleted in the MPTP-induced Parkinsonian group. The MPTP group pretreated with PYC showed significant protection of the activity of antioxidant enzymes and glutathione content when compared with the vehicle-treated MPTP group. A significantly elevated level of thiobarbituric acid reactive substances in the MPTP group was decreased significantly in the animals pretreated with PYC. An increase in the number of dopaminergic D2 receptors and decrease in the level of dopamine and its metabolite 3,4-dihydroxyphenyl acetic acid in the striatum were observed after MPTP injection, and restored significantly after PYC pretreatment. Thus, PYC may be used to prevent or reduce the deterioration caused by free radicals, thereby preventing subsequent behavioural and biochemical changes that occur in Parkinsonian mice.


Asunto(s)
Flavonoides/farmacología , Estrés Oxidativo/efectos de los fármacos , Trastornos Parkinsonianos/tratamiento farmacológico , Animales , Antioxidantes/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Flavonoides/aislamiento & purificación , Glutatión/metabolismo , Intoxicación por MPTP/tratamiento farmacológico , Intoxicación por MPTP/fisiopatología , Ratones , Trastornos Parkinsonianos/fisiopatología , Pinus/química , Extractos Vegetales/farmacología , Receptores de Dopamina D2/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
19.
Mol Neurobiol ; 57(11): 4373-4393, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32725516

RESUMEN

The neurovascular system (NVS), which consisted of neurons, glia, and vascular cells, is a functional and structural unit of the brain. The NVS regulates blood-brain barrier (BBB) permeability and cerebral blood flow (CBF), thereby maintaining the brain's microenvironment for normal functioning, neuronal survival, and information processing. Recent studies have highlighted the role of vascular dysfunction in several neurodegenerative diseases. This is not unexpected since both nervous and vascular systems are functionally interdependent and show close anatomical apposition, as well as similar molecular pathways. However, despite extensive research, the precise mechanism by which neurovascular dysfunction contributes to neurodegeneration remains incomplete. Therefore, understanding the mechanisms of neurovascular dysfunction in disease conditions may allow us to develop potent and effective therapies for prevention and treatment of neurodegenerative diseases. This review article summarizes the current research in the context of neurovascular signaling associated with neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). We also discuss the potential implication of neurovascular factor as a novel therapeutic target and prognostic marker in patients with neurodegenerative conditions. Graphical Abstract.


Asunto(s)
Sistema Nervioso/irrigación sanguínea , Enfermedades Neurodegenerativas/patología , Animales , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/fisiopatología , Circulación Cerebrovascular , Humanos , Modelos Neurológicos , Sistema Nervioso/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Enfermedades Neurodegenerativas/terapia
20.
Genes (Basel) ; 11(7)2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32629989

RESUMEN

Stroke is a leading cause of mortality and long-term disability in patients worldwide. Skeletal muscle is the primary systemic target organ of stroke that induces muscle wasting and weakness, which predominantly contribute to functional disability in stroke patients. Currently, no pharmacological drug is available to treat post-stroke muscle morbidities as the mechanisms underlying post-stroke muscle wasting remain poorly understood. To understand the stroke-mediated molecular changes occurring at the transcriptional level in skeletal muscle, the gene expression profiles and enrichment pathways were explored in a mouse model of cerebral ischemic stroke via high-throughput RNA sequencing and extensive bioinformatic analyses. RNA-seq revealed that the elevated muscle atrophy observed in response to stroke was associated with the altered expression of genes involved in proteolysis, cell cycle, extracellular matrix remodeling, and the neuromuscular junction (NMJ). These data suggest that stroke primarily targets muscle protein degradation and NMJ pathway proteins to induce muscle atrophy. Collectively, we for the first time have found a novel genome-wide transcriptome signature of post-stroke skeletal muscle in mice. Our study will provide critical information to further elucidate specific gene(s) and pathway(s) that can be targeted to mitigate accountable for post-stroke muscle atrophy and related weakness.


Asunto(s)
Infarto de la Arteria Cerebral Media/genética , Músculo Esquelético/metabolismo , Transcriptoma , Animales , Matriz Extracelular/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Ratones , Ratones Endogámicos C57BL , Unión Neuromuscular/metabolismo , Proteolisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA