Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38921973

RESUMEN

Stroke is one of the leading causes of death and long-term disabilities worldwide. In addition to interruption of blood flow, inflammation is widely recognized as an important factor mediating tissue destruction in stroke. Depending on their phenotype, microglia, the main leukocytes in the CNS, are capable of either causing further tissue damage or promoting brain restoration after stroke. ß2-integrins are cell adhesion molecules that are constitutively expressed on microglia. The function of ß2-integrins has been investigated extensively in animal models of ischemic stroke, but their role in hemorrhagic stroke is currently poorly understood. We show in this study that dysfunction of ß2-integrins is associated with improved functional outcome and decreased inflammatory cytokine expression in the brain in a mouse model of hemorrhagic stroke. Furthermore, ß2-integrins affect microglial phenotype and cytokine responses in vivo. Therefore, our findings suggest that targeting ß2-integrins in hemorrhagic stroke may be beneficial.

2.
Acta Neuropathol Commun ; 12(1): 10, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38229173

RESUMEN

Mesencephalic astrocyte-derived neurotrophic factor (MANF) has cytoprotective effects on various injuries, including cerebral ischemia, and it can promote recovery even when delivered intracranially several days after ischemic stroke. In the uninjured rodent brain, MANF protein is expressed almost exclusively in neurons, but post-ischemic MANF expression has not been characterized. We aimed to investigate how endogenous cerebral MANF protein expression evolves in infarcted human brains and rodent ischemic stroke models. During infarct progression, the cerebral MANF expression pattern both in human and rat brains shifted drastically from neurons to expression in inflammatory cells. Intense MANF immunoreactivity took place in phagocytic microglia/macrophages in the ischemic territory, peaking at two weeks post-stroke in human and one-week post-stroke in rat ischemic cortex. Using double immunofluorescence and mice lacking MANF gene and protein from neuronal stem cells, neurons, astrocytes, and oligodendrocytes, we verified that MANF expression was induced in microglia/macrophage cells in the ischemic hemisphere. Embarking on the drastic expression transition towards inflammatory cells and the impact of blood-borne inflammation in stroke, we hypothesized that exogenously delivered MANF protein can modulate tissue recovery processes. In an attempt to enhance recovery, we designed a set of proof-of-concept studies using systemic delivery of recombinant MANF in a rat model of cortical ischemic stroke. Intranasal recombinant MANF treatment decreased infarct volume and reduced the severity of neurological deficits. Intravenous recombinant MANF treatment decreased the levels of pro-inflammatory cytokines and increased the levels of anti-inflammatory cytokine IL-10 in the infarcted cortex one-day post-stroke. In conclusion, MANF protein expression is induced in activated microglia/macrophage cells in infarcted human and rodent brains, and this could implicate MANF's involvement in the regulation of post-stroke inflammation in patients and experimental animals. Moreover, systemic delivery of recombinant MANF shows promising immunomodulatory effects and therapeutic potential in experimental ischemic stroke.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Humanos , Ratas , Ratones , Animales , Accidente Cerebrovascular Isquémico/metabolismo , Ratas Sprague-Dawley , Encéfalo/metabolismo , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/uso terapéutico , Accidente Cerebrovascular/metabolismo , Infarto Cerebral/metabolismo , Inflamación/metabolismo
3.
Pharmacol Ther ; 254: 108594, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38290651

RESUMEN

Cerebral dopamine neurotrophic factor (CDNF) is an endogenous protein in humans and other vertebrates, and it has been shown to have protective and restorative effects on cells in various disease models. Although it is named as a neurotrophic factor, its actions are drastically different from classical neurotrophic factors such as neurotrophins or the glial cell line-derived neurotrophic family of proteins. Like all secreted proteins, CDNF has a signal sequence at the N-terminus, but unlike common growth factors it has a KDEL-receptor retrieval sequence at the C-terminus. Thus, CDNF is mainly located in the ER. In response to adverse effects, such as ER stress, the expression of CDNF is upregulated and can alleviate ER stress. Also different from other neurotrophic factors, CDNF reduces protein aggregation and inflammation in disease models. Although it is an ER luminal protein, it can surprisingly directly interact with alpha-synuclein, a protein involved in the pathogenesis of synucleinopathies e.g., Parkinson's disease. Pleiotropic CDNF has therapeutic potential and has been tested as a recombinant human protein and gene therapy. The neuroprotective and neurorestorative effects have been described in a number of preclinical studies of Parkinson's disease, stroke and amyotrophic lateral sclerosis. Currently, it was successfully evaluated for safety in a phase 1/2 clinical trial for Parkinson's disease. Collectively, based on recent findings on the mode of action and therapeutic potential of CDNF, its use as a drug could be expanded to other ER stress-related diseases.


Asunto(s)
Enfermedad de Parkinson , Animales , Humanos , Enfermedad de Parkinson/tratamiento farmacológico , Factores de Crecimiento Nervioso/uso terapéutico , Factores de Crecimiento Nervioso/metabolismo , Proteínas Recombinantes/uso terapéutico
4.
Biosens Bioelectron ; 241: 115579, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37690355

RESUMEN

Electrochemical sensors provide means for real-time monitoring of neurotransmitter release events, which is a relatively easy process in simple electrolytes. However, this does not apply to in vitro environments. In cell culture media, competitively adsorbing molecules are present at concentrations up to 350 000-fold excess compared to the neurotransmitter-of-interest. Because detection of dopamine and serotonin requires direct adsorption of the analyte to electrode surface, a significant loss of sensitivity occurs when recording is performed in the in vitro environment. Despite these challenges, our single-walled carbon nanotube (SWCNT) sensor was capable of selectively measuring dopamine and serotonin from cell culture medium at nanomolar concentration in real-time. A primary midbrain culture was used to prove excellent biocompatibility of our SWCNT electrodes, which is a necessity for brain-on-a-chip models. Most importantly, our sensor was able to electrochemically record spontaneous transient activity from dopaminergic cell culture without altering the culture conditions, which has not been possible earlier. Drug discovery and development requires high-throughput screening of in vitro models, being hindered by the challenges in non-invasive characterization of complex neuronal models such as organoids. Our neurotransmitter sensors could be used for real-time monitoring of complex neuronal models, providing an alternative tool for their characterization non-invasively.


Asunto(s)
Técnicas Biosensibles , Dopamina , Dopamina/química , Serotonina/metabolismo , Electrodos , Neurotransmisores/metabolismo , Técnicas Electroquímicas
5.
Prostaglandins Other Lipid Mediat ; 168: 106760, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37331425

RESUMEN

Ischemic cerebral stroke is a severe medical condition that affects about 15 million people every year and is the second leading cause of death and disability globally. Ischemic stroke results in neuronal cell death and neurological impairment. Current therapies may not adequately address the deleterious metabolic changes and may increase neurological damage. Oxygen and nutrient depletion along with the tissue damage result in endoplasmic reticulum (ER) stress, including the Unfolded Protein Response (UPR), and neuroinflammation in the affected area and cause cell death in the lesion core. The spatio-temporal production of lipid mediators, either pro-inflammatory or pro-resolving, decides the course and outcome of stroke. The modulation of the UPR as well as the resolution of inflammation promotes post-stroke cellular viability and neuroprotection. However, studies about the interplay between the UPR and bioactive lipid mediators remain elusive and this review gives insights about the crosstalk between lipid mediators and the UPR in ischemic stroke. Overall, the treatment of ischemic stroke is often inadequate due to lack of effective drugs, thus, this review will provide novel therapeutical strategies that could promote the functional recovery from ischemic stroke.


Asunto(s)
Accidente Cerebrovascular Isquémico , Humanos , Respuesta de Proteína Desplegada , Estrés del Retículo Endoplásmico , Inflamación , Lípidos
6.
Pharmacol Rev ; 75(5): 959-978, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37127349

RESUMEN

The endoplasmic reticulum (ER) is the largest organelle of the cell, composed of a continuous network of sheets and tubules, and is involved in protein, calcium (Ca2+), and lipid homeostasis. In neurons, the ER extends throughout the cell, both somal and axodendritic compartments, and is highly important for neuronal functions. A third of the proteome of a cell, secreted and membrane-bound proteins, are processed within the ER lumen and most of these proteins are vital for neuronal activity. The brain itself is high in lipid content, and many structural lipids are produced, in part, by the ER. Cholesterol and steroid synthesis are strictly regulated in the ER of the blood-brain barrier protected brain cells. The high Ca2+ level in the ER lumen and low cytosolic concentration is needed for Ca2+-based intracellular signaling, for synaptic signaling and Ca2+ waves, and for preparing proteins for correct folding in the presence of high Ca2+ concentrations to cope with the high concentrations of extracellular milieu. Particularly, ER Ca2+ is controlled in axodendritic areas for proper neurito- and synaptogenesis and synaptic plasticity and remodeling. In this review, we cover the physiologic functions of the neuronal ER and discuss it in context of common neurodegenerative diseases, focusing on pharmacological regulation of ER Ca2+ Furthermore, we postulate that heterogeneity of the ER, its protein folding capacity, and ensuring Ca2+ regulation are crucial factors for the aging and selective vulnerability of neurons in various neurodegenerative diseases. SIGNIFICANCE STATEMENT: Endoplasmic reticulum (ER) Ca2+ regulators are promising therapeutic targets for degenerative diseases for which efficacious drug therapies do not exist. The use of pharmacological probes targeting maintenance and restoration of ER Ca2+ can provide restoration of protein homeostasis (e.g., folding of complex plasma membrane signaling receptors) and slow down the degeneration process of neurons.


Asunto(s)
Enfermedades Neurodegenerativas , Humanos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Calcio de la Dieta/metabolismo , Lípidos , Señalización del Calcio
7.
Nat Neurosci ; 26(6): 1008-1020, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37169859

RESUMEN

Molecular diversity of microglia, the resident immune cells in the CNS, is reported. Whether microglial subsets characterized by the expression of specific proteins constitute subtypes with distinct functions has not been fully elucidated. Here we describe a microglial subtype expressing the enzyme arginase-1 (ARG1; that is, ARG1+ microglia) that is found predominantly in the basal forebrain and ventral striatum during early postnatal mouse development. ARG1+ microglia are enriched in phagocytic inclusions and exhibit a distinct molecular signature, including upregulation of genes such as Apoe, Clec7a, Igf1, Lgals3 and Mgl2, compared to ARG1- microglia. Microglial-specific knockdown of Arg1 results in deficient cholinergic innervation and impaired dendritic spine maturation in the hippocampus where cholinergic neurons project, which in turn results in impaired long-term potentiation and cognitive behavioral deficiencies in female mice. Our results expand on microglia diversity and provide insights into microglia subtype-specific functions.


Asunto(s)
Arginasa , Microglía , Animales , Femenino , Ratones , Arginasa/genética , Arginasa/metabolismo , Hipocampo/metabolismo , Microglía/metabolismo
8.
Cell Death Dis ; 14(2): 128, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36792604

RESUMEN

During intracerebral hemorrhage (ICH), hematoma formation at the site of blood vessel damage results in local mechanical injury. Subsequently, erythrocytes lyse to release hemoglobin and heme, which act as neurotoxins and induce inflammation and secondary brain injury, resulting in severe neurological deficits. Accelerating hematoma resorption and mitigating hematoma-induced brain edema by modulating immune cells has potential as a novel therapeutic strategy for functional recovery after ICH. Here, we show that intracerebroventricular administration of recombinant human cerebral dopamine neurotrophic factor (rhCDNF) accelerates hemorrhagic lesion resolution, reduces peri-focal edema, and improves neurological outcomes in an animal model of collagenase-induced ICH. We demonstrate that CDNF acts on microglia/macrophages in the hemorrhagic striatum by promoting scavenger receptor expression, enhancing erythrophagocytosis and increasing anti-inflammatory mediators while suppressing the production of pro-inflammatory cytokines. Administration of rhCDNF results in upregulation of the Nrf2-HO-1 pathway, but alleviation of oxidative stress and unfolded protein responses in the perihematomal area. Finally, we demonstrate that intravenous delivery of rhCDNF has beneficial effects in an animal model of ICH and that systemic application promotes scavenging by the brain's myeloid cells for the treatment of ICH.


Asunto(s)
Edema Encefálico , Lesiones Encefálicas , Animales , Humanos , Hemorragia Cerebral/complicaciones , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/patología , Inflamación/complicaciones , Hematoma/tratamiento farmacológico , Hematoma/complicaciones , Hematoma/metabolismo , Inmunidad Innata , Modelos Animales de Enfermedad , Edema Encefálico/complicaciones , Factores de Crecimiento Nervioso/uso terapéutico
9.
Biochem Soc Trans ; 51(1): 245-257, 2023 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-36794783

RESUMEN

Synucleinopathies constitute a disease family named after alpha-synuclein protein, which is a significant component of the intracellular inclusions called Lewy bodies. Accompanying the progressive neurodegeneration, Lewy bodies and neurites are the main histopathologies of synucleinopathies. The complicated role of alpha-synuclein in the disease pathology makes it an attractive therapeutic target for disease-modifying treatments. GDNF is one of the most potent neurotrophic factors for dopamine neurons, whereas CDNF is protective and neurorestorative with entirely different mechanisms of action. Both have been in the clinical trials for the most common synucleinopathy, Parkinson's disease. With the AAV-GDNF clinical trials ongoing and the CDNF trial being finalized, their effects on abnormal alpha-synuclein accumulation are of great interest. Previous animal studies with an alpha-synuclein overexpression model have shown that GDNF was ineffective against alpha-synuclein accumulation. However, a recent study with cell culture and animal models of alpha-synuclein fibril inoculation has demonstrated the opposite by revealing that the GDNF/RET signaling cascade is required for the protective effect of GDNF on alpha-synuclein aggregation. CDNF, an ER resident protein, was shown to bind alpha-synuclein directly. CDNF reduced the uptake of alpha-synuclein fibrils by the neurons and alleviated the behavioral deficits induced by fibrils injected into the mouse brain. Thus, GDNF and CDNF can modulate different symptoms and pathologies of Parkinson's disease, and perhaps, similarly for other synucleinopathies. Their unique mechanisms for preventing alpha-synuclein-related pathology should be studied more carefully to develop disease-modifying therapies.


Asunto(s)
Enfermedad de Parkinson , Sinucleinopatías , Animales , Ratones , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Dopamina/metabolismo , Dopamina/farmacología , Dopamina/uso terapéutico , Sinucleinopatías/metabolismo , Sinucleinopatías/patología , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Neuronas Dopaminérgicas/metabolismo
10.
Front Cell Neurosci ; 16: 900725, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35783104

RESUMEN

Stroke is a devastating medical condition with no treatment to hasten recovery. Its abrupt nature results in cataclysmic changes in the affected tissues. Resident cells fail to cope with the cellular stress resulting in massive cell death, which cannot be endogenously repaired. A potential strategy to improve stroke outcomes is to boost endogenous pro-survival pathways. The unfolded protein response (UPR), an evolutionarily conserved stress response, provides a promising opportunity to ameliorate the survival of stressed cells. Recent studies from us and others have pointed toward mesencephalic astrocyte-derived neurotrophic factor (MANF) being a UPR responsive gene with an active role in maintaining proteostasis. Its pro-survival effects have been demonstrated in several disease models such as diabetes, neurodegeneration, and stroke. MANF has an ER-signal peptide and an ER-retention signal; it is secreted by ER calcium depletion and exits cells upon cell death. Although its functions remain elusive, conducted experiments suggest that the endogenous MANF in the ER lumen and exogenously administered MANF protein have different mechanisms of action. Here, we will revisit recent and older bodies of literature aiming to delineate the expression profile of MANF. We will focus on its neuroprotective roles in regulating neurogenesis and inflammation upon post-stroke administration. At the same time, we will investigate commonalities and differences with another UPR responsive gene, X-box binding protein 1 (XBP1), which has recently been associated with MANF's function. This will be the first systematic comparison of these two UPR responsive genes aiming at revealing previously uncovered associations between them. Overall, understanding the mode of action of these UPR responsive genes could provide novel approaches to promote cell survival.

11.
Biomedicines ; 10(6)2022 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-35740467

RESUMEN

Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in substantia nigra pars compacta, which leads to the motor control deficits. Recently, cell transplantation is a cutting-edge technique for the therapy of PD. Nevertheless, one key bottleneck to realizing such potential is allogenic immune reaction of tissue grafts by recipients. Cerebral dopamine neurotrophic factor (CDNF) was shown to possess immune-modulatory properties that benefit neurodegenerative diseases. We hypothesized that co-administration of CDNF with fetal ventral mesencephalic (VM) tissue can improve the success of VM replacement therapies by attenuating immune responses. Hemiparkinsonian rats were generated by injecting 6-hydroxydopamine (6-OHDA) into the right medial forebrain bundle of Sprague Dawley (SD) rats. The rats were then intrastriatally transplanted with VM tissue from rats, with/without CDNF administration. Recovery of dopaminergic function and survival of the grafts were evaluated using the apomorphine-induced rotation test and small-animal positron emission tomography (PET) coupled with [18F] DOPA or [18F] FE-PE2I, respectively. In addition, transplantation-related inflammatory response was determined by uptake of [18F] FEPPA in the grafted side of striatum. Immunohistochemistry (IHC) examination was used to determine the survival of the grated dopaminergic neurons in the striatum and to investigate immune-modulatory effects of CDNF. The modulation of inflammatory responses caused by CDNF might involve enhancing M2 subset polarization and increasing fractal dimensions of 6-OHDA-treated BV2 microglial cell line. Analysis of CDNF-induced changes to gene expressions of 6-OHDA-stimulated BV2 cells implies that these alternations of the biomarkers and microglial morphology are implicated in the upregulation of protein kinase B signaling as well as regulation of catalytic, transferase, and protein serine/threonine kinase activity. The effects of CDNF on 6-OHDA-induced alternation of the canonical pathway in BV2 microglial cells is highly associated with PI3K-mediated phagosome formation. Our results are the first to show that CDNF administration enhances the survival of the grafted dopaminergic neurons and improves functional recovery in PD animal model. Modulation of the polarization, morphological characteristics, and transcriptional profiles of 6-OHDA-stimualted microglia by CDNF may possess these properties in transplantation-based regenerative therapies.

12.
Acta Biomater ; 146: 235-247, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35487425

RESUMEN

Vertically aligned carbon nanofibers (VACNFs) are promising material candidates for neural biosensors due to their ability to detect neurotransmitters in physiological concentrations. However, the expected high rigidity of CNFs could induce mechanical mismatch with the brain tissue, eliciting formation of a glial scar around the electrode and thus loss of functionality. We have evaluated mechanical biocompatibility of VACNFs by growing nickel-catalyzed carbon nanofibers of different lengths and inter-fiber distances. Long nanofibers with large inter-fiber distance prevented maturation of focal adhesions, thus constraining cells from obtaining a highly spread morphology that is observed when astrocytes are being contacted with stiff materials commonly used in neural implants. A silicon nanopillar array with 500 nm inter-pillar distance was used to reveal that this inhibition of focal adhesion maturation occurs due to the surface nanoscale geometry, more precisely the inter-fiber distance. Live cell atomic force microscopy was used to confirm astrocytes being significantly softer on the long Ni-CNFs compared to other surfaces, including a soft gelatin hydrogel. We also observed hippocampal neurons to mature and form synaptic contacts when being cultured on both long and short carbon nanofibers, without having to use any adhesive proteins or a glial monoculture, indicating high cytocompatibility of the material also with neuronal population. In contrast, neurons cultured on a planar tetrahedral amorphous carbon sample showed immature neurites and indications of early-stage apoptosis. Our results demonstrate that mechanical biocompatibility of biomaterials is greatly affected by their nanoscale surface geometry, which provides means for controlling how the materials and their mechanical properties are perceived by the cells. STATEMENT OF SIGNIFICANCE: Our research article shows, how nanoscale surface geometry determines mechanical biocompatibility of apparently stiff materials. Specifically, astrocytes were prevented from obtaining highly spread morphology when their adhesion site maturation was inhibited, showing similar morphology on nominally stiff vertically aligned carbon fiber (VACNF) substrates as when being cultured on ultrasoft surfaces. Furthermore, hippocampal neurons matured well and formed synapses on these carbon nanofibers, indicating high biocompatibility of the materials. Interestingly, the same VACNF materials that were used in this study have earlier also been proven to be capable for electrophysiological recordings and sensing neurotransmitters at physiological concentrations with ultra-high sensitivity and selectivity, thus providing a platform for future neural probes or smart culturing surfaces with superior sensing performance and biocompatibility.


Asunto(s)
Nanofibras , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Carbono/química , Electrodos , Nanofibras/química , Neuritas
13.
Biomolecules ; 12(4)2022 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-35454152

RESUMEN

There are several links between insulin resistance and neurodegenerative disorders such as Parkinson's disease. However, the direct influence of insulin signaling on abnormal α-synuclein accumulation-a hallmark of Parkinson's disease-remains poorly explored. To our best knowledge, this work is the first attempt to investigate the direct effects of insulin signaling on pathological α-synuclein accumulation induced by the addition of α-synuclein preformed fibrils in primary dopaminergic neurons. We found that modifying insulin signaling through (1) insulin receptor inhibitor GSK1904529A, (2) SHIP2 inhibitor AS1949490 or (3) PTEN inhibitor VO-OHpic failed to significantly affect α-synuclein aggregation in dopaminergic neurons, in contrast to the aggregation-reducing effects observed after the addition of glial cell line-derived neurotrophic factor. Subsequently, we tested different media formulations, with and without insulin. Again, removal of insulin from cell culturing media showed no effect on α-synuclein accumulation. We observed, however, a reduced α-synuclein aggregation in neurons cultured in neurobasal medium with a B27 supplement, regardless of the presence of insulin, in contrast to DMEM/F12 medium with an N2 supplement. The effects of culture conditions were present only in dopaminergic but not in primary cortical or hippocampal cells, indicating the unique sensitivity of the former. Altogether, our data contravene the direct involvement of insulin signaling in the modulation of α-synuclein aggregation in dopamine neurons. Moreover, we show that the choice of culturing media can significantly affect preformed fibril-induced α-synuclein phosphorylation in a primary dopaminergic cell culture.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Técnicas de Cultivo de Célula , Dopamina , Neuronas Dopaminérgicas , Humanos , Insulina/farmacología , Enfermedad de Parkinson/patología
14.
J Control Release ; 344: 214-224, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35301056

RESUMEN

The blood-brain barrier significantly limits effective drug delivery to central nervous system (CNS) targets. The recently characterized glymphatic system offers a perivascular highway for intrathecally (i.t.) administered drugs to reach deep brain structures. Although periarterial cerebrospinal fluid (CSF) influx and concomitant brain drug delivery can be enhanced by pharmacological or hyperosmotic interventions, their effects on drug delivery to the spinal cord, an important target for many drugs, have not been addressed. Hence, we studied in rats whether enhancement of periarterial flow by systemic hypertonic solution might be utilized to enhance spinal delivery and efficacy of i.t. morphine. We also studied whether the hyperosmolar intervention affects brain or cerebrospinal fluid drug concentrations after systemic administration. Periarterial CSF influx was enhanced by intraperitoneal injection of hypertonic saline (HTS, 5.8%, 20 ml/kg, 40 mOsm/kg). The antinociceptive effects of morphine were characterized, using tail flick, hot plate and paw pressure tests. Drug concentrations in serum, tissue and microdialysis samples were determined by liquid chromatography-tandem mass spectrometry. Compared with isotonic solution, HTS increased concentrations of spinal i.t. administered morphine by 240% at the administration level (T13-L1) at 60 min and increased the antinociceptive effect of morphine in tail flick, hot plate, and paw pressure tests. HTS also independently increased hot plate and paw pressure latencies but had no effect in the tail flick test. HTS transiently increased the penetration of intravenous morphine into the lateral ventricle, but not into the hippocampus. In conclusion, acute systemic hyperosmolality is a promising intervention for enhanced spinal delivery of i.t. administered morphine. The relevance of this intervention should be expanded to other i.t. drugs and brought to clinical trials.


Asunto(s)
Morfina , Médula Espinal , Animales , Inyecciones Espinales , Dimensión del Dolor , Ratas , Ratas Sprague-Dawley
15.
Bioorg Med Chem Lett ; 64: 128677, 2022 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-35301136

RESUMEN

Preventing the aggregation of certain amyloid proteins has the potential to slow down the progression of diseases like Alzheimer's, Parkinson's, and type 2 diabetes mellitus. During a high-throughput screen of 300 Australian marine invertebrate extracts, the extract of the marine sponge Thorectandra sp. 4408 displayed binding activity to the Parkinson's disease-associated protein, α-synuclein. Isolation of the active component led to its identification as the known plant growth promoter asterubine (1). This molecule shares distinct structural similarities with potent amyloid beta aggregation inhibitors tramiprosate (homotaurine) and ALZ-801. Herein we report the isolation, NMR data acquired in DMSO and α-synuclein binding activity of asterubine (1).


Asunto(s)
Diabetes Mellitus Tipo 2 , Enfermedad de Parkinson , Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Australia , Humanos , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína
16.
Eur J Hum Genet ; 30(5): 619-627, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35087184

RESUMEN

Biallelic loss-of-function variants in the SMG9 gene, encoding a regulatory subunit of the mRNA nonsense-mediated decay (NMD) machinery, are reported to cause heart and brain malformation syndrome. Here we report five patients from three unrelated families with intellectual disability (ID) and a novel pathogenic SMG9 c.551 T > C p.(Val184Ala) homozygous missense variant, identified using exome sequencing. Sanger sequencing confirmed recessive segregation in each family. SMG9 c.551T > C p.(Val184Ala) is most likely an autozygous variant identical by descent. Characteristic clinical findings in patients were mild to moderate ID, intention tremor, pyramidal signs, dyspraxia, and ocular manifestations. We used RNA sequencing of patients and age- and sex-matched healthy controls to assess the effect of the variant. RNA sequencing revealed that the SMG9 c.551T > C variant did not affect the splicing or expression level of SMG9 gene products, and allele-specific expression analysis did not provide evidence that the nonsense mRNA-induced NMD was affected. Differential gene expression analysis identified prevalent upregulation of genes in patients, including the genes SMOX, OSBP2, GPX3, and ZNF155. These findings suggest that normal SMG9 function may be involved in transcriptional regulation without affecting nonsense mRNA-induced NMD. In conclusion, we demonstrate that the SMG9 c.551T > C missense variant causes a neurodevelopmental disorder and impacts gene expression. NMD components have roles beyond aberrant mRNA degradation that are crucial for neurocognitive development.


Asunto(s)
Discapacidad Intelectual , Péptidos y Proteínas de Señalización Intracelular , Degradación de ARNm Mediada por Codón sin Sentido , Alelos , Homocigoto , Humanos , Discapacidad Intelectual/genética , Péptidos y Proteínas de Señalización Intracelular/genética , ARN Mensajero/genética
17.
Alcohol Alcohol ; 57(4): 405-412, 2022 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-33893472

RESUMEN

AIMS: Recently we developed a model to study alcohol-seeking behaviour after withdrawal in a social context in female mice. The model raised several questions that we were eager to address to improve methodology. METHODS: In our model, female mice were group-housed in automated cages with three conditioned (CS+) corners and water in both sides of one separate non-conditioned corner. Water was available with opened doors at all the time of training. We established conditioning by pairing alcohol drinking with light cues. Here, we introduced prolonged access to increasing concentrations of alcohol instead of intermittent access. To study motivation to drink alcohol, we carried out the extinction tests on withdrawal days 1 (WD1) and 10 (WD10). During tests, the light cues were present in conditioned corners, but there was no liquid in the bottles. RESULTS: We found that the number of visits and nosepokes in the CS+ corner in the alcohol group was much higher than in the water group. Also, during training, the consumption of alcohol was increasing. In the extinction tests, we found that the number of nosepokes in the CS+ corner increased in the alcohol group on both WD1 and WD10. CONCLUSIONS: Our study supports that alcohol-seeking behaviour after withdrawal can be modelled and studied in group-housed animals and environments without social isolation.


Asunto(s)
Consumo de Bebidas Alcohólicas , Etanol , Animales , Señales (Psicología) , Femenino , Ratones , Ratones Endogámicos C57BL , Medio Social , Agua
18.
J Biomed Sci ; 28(1): 87, 2021 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-34923968

RESUMEN

BACKGROUND: Craving for alcohol, in other words powerful desire to drink after withdrawal, is an important contributor to the development and maintenance of alcoholism. Here, we studied the role of GDNF (glial cell line-derived neurotrophic factor) and BDNF (brain-derived neurotrophic factor) on alcohol-seeking behavior in group-housed female mice. METHODS: We modeled alcohol-seeking behavior in C57Bl/6J female mice. The behavioral experiments in group-housed female mice were performed in an automated IntelliCage system. We conducted RT-qPCR analysis of Gdnf, Bdnf, Manf and Cdnf expression in different areas of the female mouse brain after alcohol drinking conditioning. We injected an adeno-associated virus (AAV) vector expressing human GDNF or BDNF in mouse nucleus accumbens (NAc) after ten days of alcohol drinking conditioning and assessed alcohol-seeking behavior. Behavioral data were analyzed by two-way repeated-measures ANOVA, and statistically significant effects were followed by Bonferroni's post hoc test. The student's t-test was used to analyze qPCR data. RESULTS: The RT-qPCR data showed that Gdnf mRNA level in NAc was more than four times higher (p < 0.0001) in the mice from the sweetened alcohol group compared to the water group. Our data showed a more than a two-fold decrease in Manf mRNA (p = 0.04) and Cdnf mRNA (p = 0.02) levels in the hippocampus and Manf mRNA in the VTA (p = 0.04) after alcohol consumption. Two-fold endogenous overexpression of Gdnf mRNA and lack of CDNF did not affect alcohol-seeking behavior. The AVV-GDNF overexpression in nucleus accumbens suppressed alcohol-seeking behavior while overexpression of BDNF did not. CONCLUSIONS: The effect of increased endogenous Gdnf mRNA level in female mice upon alcohol drinking has remained unknown. Our data suggest that an increase in endogenous GDNF expression upon alcohol drinking occurs in response to the activation of another mesolimbic reward pathway participant.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Ansia , Regulación de la Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Núcleo Accumbens/metabolismo , Animales , Femenino , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Ratones , Ratones Endogámicos C57BL , Conducta Social
19.
ACS Chem Neurosci ; 12(13): 2273-2279, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34110772

RESUMEN

Neurodegenerative diseases are associated with failed proteostasis and accumulation of insoluble protein aggregates that compromise neuronal function and survival. In Parkinson's disease, a major pathological finding is Lewy bodies and neurites that are mainly composed of phosphorylated and aggregated α-synuclein and fragments of organelle membranes. Here, we analyzed a series of selective inhibitors acting on multidomain proteins CBP and p300 that contain both lysine acetyltransferase and bromodomains and are responsible for the recognition and enzymatic modification of lysine residues. By using high-affinity inhibitors, A-485, GNE-049, and SGC-CBP30, we explored the role of two closely related proteins, CBP and p300, as promising targets for selective attenuation of α-synuclein aggregation. Our data show that selective CBP/p300 inhibitors may alter the course of pathological α-synuclein accumulation in primary mouse embryonic dopaminergic neurons. Hence, drug-like CBP/p300 inhibitors provide an effective approach for the development of high-affinity drug candidates preventing α-synuclein aggregation via systemic administration.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Animales , Neuronas Dopaminérgicas , Cuerpos de Lewy , Ratones , Dominios Proteicos
20.
Mol Ther ; 29(9): 2821-2840, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33940158

RESUMEN

A molecular hallmark in Parkinson's disease (PD) pathogenesis are α-synuclein aggregates. Cerebral dopamine neurotrophic factor (CDNF) is an atypical growth factor that is mostly resident in the endoplasmic reticulum but exerts its effects both intracellularly and extracellularly. One of the beneficial effects of CDNF can be protecting neurons from the toxic effects of α-synuclein. Here, we investigated the effects of CDNF on α-synuclein aggregation in vitro and in vivo. We found that CDNF directly interacts with α-synuclein with a KD = 23 ± 6 nM and reduces its auto-association. Using nuclear magnetic resonance (NMR) spectroscopy, we identified interaction sites on the CDNF protein. Remarkably, CDNF reduces the neuronal internalization of α-synuclein fibrils and induces the formation of insoluble phosphorylated α-synuclein inclusions. Intra-striatal CDNF administration alleviates motor deficits in rodents challenged with α-synuclein fibrils, though it did not reduce the number of phosphorylated α-synuclein inclusions in the substantia nigra. CDNF's beneficial effects on rodent behavior appear not to be related to the number of inclusions formed in the current context, and further study of its effects on the aggregation mechanism in vivo are needed. Nonetheless, the interaction of CDNF with α-synuclein, modifying its aggregation, spreading, and associated behavioral alterations, provides novel insights into the potential of CDNF as a therapeutic strategy in PD and other synucleinopathies.


Asunto(s)
Factores de Crecimiento Nervioso/química , Factores de Crecimiento Nervioso/metabolismo , Enfermedad de Parkinson/fisiopatología , Sustancia Negra/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Animales , Sitios de Unión , Línea Celular , Modelos Animales de Enfermedad , Dopamina/metabolismo , Humanos , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Modelos Moleculares , Enfermedad de Parkinson/metabolismo , Fosforilación , Cultivo Primario de Células , Agregado de Proteínas , Unión Proteica , Conformación Proteica , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA