Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Cells ; 13(10)2024 May 16.
Article in English | MEDLINE | ID: mdl-38786067

ABSTRACT

This systematic review aims to gather evidence on the mechanisms triggered by diverse preconditioning strategies for mesenchymal stem cells (MSCs) and their impact on their potential to treat ischemic and traumatic injuries affecting the nervous system. The 52 studies included in this review report nine different types of preconditioning, namely, manipulation of oxygen pressure, exposure to chemical substances, lesion mediators or inflammatory factors, usage of ultrasound, magnetic fields or biomechanical forces, and culture in scaffolds or 3D cultures. All these preconditioning strategies were reported to interfere with cellular pathways that influence MSCs' survival and migration, alter MSCs' phenotype, and modulate the secretome and proteome of these cells, among others. The effects on MSCs' phenotype and characteristics influenced MSCs' performance in models of injury, namely by increasing the homing and integration of the cells in the lesioned area and inducing the secretion of growth factors and cytokines. The administration of preconditioned MSCs promoted tissue regeneration, reduced neuroinflammation, and increased angiogenesis and myelinization in rodent models of stroke, traumatic brain injury, and spinal cord injury. These effects were also translated into improved cognitive and motor functions, suggesting an increased therapeutic potential of MSCs after preconditioning. Importantly, none of the studies reported adverse effects or less therapeutic potential with these strategies. Overall, we can conclude that all the preconditioning strategies included in this review can stimulate pathways that relate to the therapeutic effects of MSCs. Thus, it would be interesting to explore whether combining different preconditioning strategies can further boost the reparative effects of MSCs, solving some limitations of MSCs' therapy, namely donor-associated variability.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Animals , Mesenchymal Stem Cell Transplantation/methods , Nervous System Diseases/therapy
2.
J Neuroimmunol ; 387: 578273, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38183948

ABSTRACT

Neuroinflammation is a pathological mechanism contributing to neurodegenerative diseases. For in-depth studies of neuroinflammation, several animal models reported reproducing behavioral dysfunctions and cellular pathological mechanisms induced by brain inflammation. One of the most popular models of neuroinflammation is the one generated by lipopolysaccharide exposure. Despite its importance, the reported results using this model show high heterogeneity, making it difficult to analyze and compare the outcomes between studies. Therefore, the current review aims to summarize the different experimental paradigms used to reproduce neuroinflammation by lipopolysaccharide exposure and its respective outcomes, helping to choose the model that better suits each specific research aim.


Subject(s)
Inflammation , Neuroinflammatory Diseases , Animals , Inflammation/chemically induced , Inflammation/pathology , Lipopolysaccharides/toxicity , Microglia/pathology , Disease Models, Animal
3.
Int J Mol Sci ; 24(22)2023 Nov 20.
Article in English | MEDLINE | ID: mdl-38003733

ABSTRACT

Due to their characteristics, mesenchymal stem cells (MSCs) are considered a potential therapy for brain tissue injury or degeneration. Nevertheless, despite the promising results observed, there has been a growing interest in the use of cell-free therapies in regenerative medicine, such as the use of stem cell secretome. This review provides an in-depth compilation of data regarding the secretome composition, protocols used for its preparation, as well as existing information on the impact of secretome administration on various brain conditions, pointing out gaps and highlighting relevant findings. Moreover, due to the ability of MSCs to respond differently depending on their microenvironment, preconditioning of MSCs has been used to modulate their composition and, consequently, their therapeutic potential. The different strategies used to modulate the MSC secretome were also reviewed. Although secretome administration was effective in improving functional impairments, regeneration, neuroprotection, and reducing inflammation in brain tissue, a high variability in secretome preparation and administration was identified, compromising the transposition of preclinical data to clinical studies. Indeed, there are no reports of the use of secretome in clinical trials. Despite the existing limitations and lack of clinical data, secretome administration is a potential tool for the treatment of various diseases that impact the CNS.


Subject(s)
Brain Injuries , Mesenchymal Stem Cells , Humans , Secretome , Regenerative Medicine/methods , Cell- and Tissue-Based Therapy
4.
Transl Psychiatry ; 13(1): 312, 2023 Oct 06.
Article in English | MEDLINE | ID: mdl-37803004

ABSTRACT

Striatal dysfunction has been implicated in the pathophysiology of schizophrenia, a disorder characterized by positive symptoms such as hallucinations and delusions. Haloperidol is a typical antipsychotic medication used in the treatment of schizophrenia that is known to antagonize dopamine D2 receptors, which are abundantly expressed in the striatum. However, haloperidol's delayed therapeutic effect also suggests a mechanism of action that may go beyond the acute blocking of D2 receptors. Here, we performed proteomic analysis of striatum brain tissue and found more than 400 proteins significantly altered after 30 days of chronic haloperidol treatment in mice, namely proteins involved in glutamatergic and GABAergic synaptic transmission. Cell-type specific electrophysiological recordings further revealed that haloperidol not only reduces the excitability of striatal medium spiny neurons expressing dopamine D2 receptors (D2-MSNs) but also affects D1-MSNs by increasing the ratio of inhibitory/excitatory synaptic transmission (I/E ratio) specifically onto D1-MSNs but not D2-MSNs. Therefore, we propose the slow remodeling of D1-MSNs as a mechanism mediating the delayed therapeutic effect of haloperidol over striatum circuits. Understanding how haloperidol exactly contributes to treating schizophrenia symptoms may help to improve therapeutic outcomes and elucidate the molecular underpinnings of this disorder.


Subject(s)
Antipsychotic Agents , Haloperidol , Mice , Animals , Haloperidol/pharmacology , Proteomics , Neurons/metabolism , Corpus Striatum/metabolism , Antipsychotic Agents/pharmacology , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D1 , Mice, Transgenic
5.
J Neurochem ; 164(6): 813-828, 2023 03.
Article in English | MEDLINE | ID: mdl-36477745

ABSTRACT

Due to its ability to improve the most frequent clinical sequelae left by ischemia, repetitive transcranial magnetic stimulation has been considered a promising therapeutic strategy for stroke. Those improvements are associated with changes in neurons and their synaptic liaisons. However, the hypothesis that this technique modulates astrocytes, potentiating their neuroprotective capabilities, was also raised. This study aims to identify the effects triggered by high-frequency repetitive magnetic stimulation (HF-rMS) on astrocytes that contribute to its neuroprotective effects. Neuron-glia and astrocyte cortical cultures subject to oxygen and glucose deprivation were used as an in vitro model of ischemia. Neuroprotection promoted by HF-rMS was evaluated by analysis of markers of neuronal activity and morphometric analysis of neurons. Glial reactivity was determined by immunocytochemistry. The levels of growth factors in the astrocyte-conditioned medium (CM) were assessed through a Growth Factor Array and glial-derived neurotrophic factor (GDNF) expression was analyzed by RT-PCR and Western blot. Our results show that neurons injured by ischemia can be rescued through the modulation of astrocytes by HF-rMS. This modulation helps to maintain the number and length of neurites and increases the number of neurons expressing ERK1/2 and c-Fos. Analysis of the astrocyte-CM showed that HF-rMS stimulated the release of several trophic factors by astrocytes. Moreover, GDNF was one of the released factors that contributed to the recovery mechanisms triggered by HF-rMS. Our results show that modulation of astrocytes by HF-rMS effectively rescues neurons injured by ischemia and suggest that by targeting astrocytes this approach can also be used to promote neuroprotection in other brain lesions.


Subject(s)
Astrocytes , Glial Cell Line-Derived Neurotrophic Factor , Humans , Astrocytes/metabolism , Secretome , Cells, Cultured , Neurons/metabolism , Ischemia/metabolism , Intercellular Signaling Peptides and Proteins , Magnetic Phenomena
6.
Cells ; 13(1)2023 12 29.
Article in English | MEDLINE | ID: mdl-38201277

ABSTRACT

Overactivation of microglial cells seems to play a crucial role in the degeneration of dopaminergic neurons occurring in Parkinson's disease. We have previously demonstrated that glial cell line-derived neurotrophic factor (GDNF) present in astrocytes secretome modulates microglial responses induced by an inflammatory insult. Therefore, astrocyte-derived soluble factors may include relevant molecular players of therapeutic interest in the control of excessive neuroinflammatory responses. However, in vivo, the control of neuroinflammation is more complex as it depends on the interaction between different types of cells other than microglia and astrocytes. Whether neurons may interfere in the astrocyte-microglia crosstalk, affecting the control of microglial reactivity exerted by astrocytes, is unclear. Therefore, the present work aimed to disclose if the control of microglial responses mediated by astrocyte-derived factors, including GDNF, could be affected by the crosstalk with neurons, impacting GDNF's ability to protect dopaminergic neurons exposed to a pro-inflammatory environment. Also, we aimed to disclose if the protection of dopaminergic neurons by GDNF involves the modulation of microglial cells. Our results show that the neuroprotective effect of GDNF was mediated, at least in part, by a direct action on microglial cells through the GDNF family receptor α-1. However, this protective effect seems to be impaired by other mediators released in response to the neuron-astrocyte crosstalk since neuron-astrocyte secretome, in contrast to astrocytes secretome, was unable to protect dopaminergic neurons from the injury triggered by lipopolysaccharide-activated microglia. Supplementation with exogenous GDNF was needed to afford protection of dopaminergic neurons exposed to the inflammatory environment. In conclusion, our results revealed that dopaminergic protective effects promoted by GDNF involve the control of microglial reactivity. However, endogenous GDNF is insufficient to confer dopaminergic neuron protection against an inflammatory insult. This reinforces the importance of further developing new therapeutic strategies aiming at providing GDNF or enhancing its expression in the brain regions affected by Parkinson's disease.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor , Parkinson Disease , Humans , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Microglia , Dopamine , Dopaminergic Neurons
7.
Cell Mol Neurobiol ; 42(7): 2289-2304, 2022 Oct.
Article in English | MEDLINE | ID: mdl-34032948

ABSTRACT

The Neurovascular Unit (NVU) is formed by vascular and neural cells controlling the cerebral hyperaemia. All the components are anatomically and functionally linked to each other, resulting in a highly efficient regulation of the cerebral blood flow, which, when interrupted, can lead to stroke. An ischemic stroke (IS) is the most common type of stroke with high rates of morbidity, mortality and disability. Therefore, it is of extreme importance to protect the functional and structural integrity of the NVU in patients with IS, understanding the mechanisms involved and how it affects each component of the NVU. Thus, the aim of this work is to analyse how the vascular smooth muscle cells from the rat middle cerebral artery function/react after an ischemic event. To mimic this event, primary cortical cultures were challenged to oxygen and glucose deprivation (OGD) for 4 h and 6 h, and the smooth muscle cells (SMCs) contractility was analysed after exposure to different media previously conditioned by the cortical cultures upon reperfusion. The results show a dual effect on the SMCs response to the vasorelaxant agent, only for cells exposed to the reperfusion media conditioned by neuron-glia cultures challenged by OGD, leading to increased relaxation of the SMCs for OGD 4 h, whereas for OGD 6 h the effect is reversed leading to contraction of the SMCs. These differences demonstrate that the astrocytes mediate the vasoactive response of vascular smooth muscle by releasing factors into the reperfusion medium, and the hypoxia time is fundamental for a beneficial/harmful response by the vascular smooth muscle.


Subject(s)
Brain Ischemia , Ischemic Stroke , Reperfusion Injury , Stroke , Animals , Cells, Cultured , Glucose , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Oxygen , Rats
8.
Int J Mol Sci ; 24(1)2022 Dec 23.
Article in English | MEDLINE | ID: mdl-36613698

ABSTRACT

Neonatal hypoxic-ischemic encephalopathy (HIE) is one of the leading causes of death and long-term disability in the perinatal period. Currently, therapeutic hypothermia is the standard of care for this condition with modest efficacy and strict enrollment criteria. Therapy with umbilical cord blood cells (UCBC) has come forward as a strong candidate for the treatment of neonatal HIE, but no preclinical studies have yet compared the action of UCBC combined with hypothermia (HT) with the action of each therapy by itself. Thus, to evaluate the potential of each therapeutic approach, a hypoxic-ischemic brain lesion was induced in postnatal day ten rat pups; two hours later, HT was applied for 4 h; and 24, 48, and 72 h post-injury, UCBC were administered intravenously. The neonatal hypoxic-ischemic injury led to a brain lesion involving about 48% of the left hemisphere that was not improved by HT (36%) or UCBC alone (28%), but only with the combined therapies (25%; p = 0.0294). Moreover, a decrease in glial reactivity and improved functional outcomes were observed in both groups treated with UCBC. Overall, these results support UCBC as a successful therapeutic approach for HIE, even when treatment with therapeutic hypothermia is not possible.


Subject(s)
Hypothermia, Induced , Hypothermia , Hypoxia-Ischemia, Brain , Rats , Animals , Neuroprotection , Fetal Blood , Hypoxia-Ischemia, Brain/therapy , Hypoxia-Ischemia, Brain/pathology , Ischemia/therapy
9.
Int J Mol Sci ; 22(6)2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33808671

ABSTRACT

Neonatal hypoxic-ischemic encephalopathy (HIE) is an important cause of mortality and morbidity in the perinatal period. This condition results from a period of ischemia and hypoxia to the brain of neonates, leading to several disorders that profoundly affect the daily life of patients and their families. Currently, therapeutic hypothermia (TH) is the standard of care in developing countries; however, TH is not always effective, especially in severe cases of HIE. Addressing this concern, several preclinical studies assessed the potential of stem cell therapy (SCT) for HIE. With this systematic review, we gathered information included in 58 preclinical studies from the last decade, focusing on the ones using stem cells isolated from the umbilical cord blood, umbilical cord tissue, placenta, and bone marrow. Outstandingly, about 80% of these studies reported a significant improvement of cognitive and/or sensorimotor function, as well as decreased brain damage. These results show the potential of SCT for HIE and the possibility of this therapy, in combination with TH, becoming the next therapeutic approach for HIE. Nonetheless, few preclinical studies assessed the combination of TH and SCT for HIE, and the existent studies show some contradictory results, revealing the need to further explore this line of research.


Subject(s)
Brain Diseases/etiology , Brain Diseases/therapy , Cell- and Tissue-Based Therapy , Hypoxia-Ischemia, Brain/complications , Stem Cell Transplantation , Animals , Astrocytes , Brain Diseases/metabolism , Brain Diseases/pathology , Cell Differentiation , Cord Blood Stem Cell Transplantation , Disease Models, Animal , Humans , Hypothermia, Induced , Infant, Newborn , Mesenchymal Stem Cell Transplantation , Microglia , Neurogenesis , Neurons , Oxidative Stress , Standard of Care , Stem Cell Transplantation/methods
10.
J Neurosci Res ; 99(5): 1414-1432, 2021 05.
Article in English | MEDLINE | ID: mdl-33522025

ABSTRACT

After decades of effort, there are no effective clinical treatments to induce the recovery of ischemia-injured tissues, and among the several strategies that have been explored, repetitive transcranial magnetic stimulation has proven to be one of the most promising, with beneficial effects in limb motor function, aphasia, hemispatial neglect, or dysphagia. Despite the clinical evidences, little is known about the mechanisms underlying those effects. The present study aimed to explore the cellular and molecular effects of high-frequency repetitive magnetic stimulation (HF-rMS) on an in vitro model of ischemia. Using primary cortical cultures exposed to oxygen and glucose deprivation followed by reperfusion, we observed that HF-rMS treatment prevents the ischemia-induced neuronal death by 21.2%, and the neurite degeneration triggered by ischemia. Our results also demonstrate that with this treatment there is an increase of 89.2% on the number cells expressing ERK1/2, of 20.1% on the number of cells expressing c-Fos, and a synaptogenic effect, through an increase of 62.9% in the number of synaptic puncta as well as of 49.4% in their intensity. Interestingly, our results indicate that astrocytes are crucial to the beneficial effects triggered by HF-rMS after ischemia, thus suggesting a direct effect of HF-rMS on these cells. The modulation of astrocytes with this non-invasive brain stimulation technique is a promising approach to promote the recovery of ischemia-induced injured tissues; however, it is essential to understand how these effects can be modulated in order to optimize the protocols and enhance the beneficial outcomes.


Subject(s)
Astrocytes/physiology , Brain Ischemia/pathology , Cerebral Cortex/physiology , Neurons/physiology , Recovery of Function/physiology , Transcranial Magnetic Stimulation/methods , Animals , Brain Ischemia/therapy , Cells, Cultured , Cerebral Cortex/cytology , Female , Male , Pregnancy , Rats , Rats, Wistar , Stroke/pathology , Stroke/therapy
11.
J Vis Exp ; (165)2020 11 14.
Article in English | MEDLINE | ID: mdl-33252110

ABSTRACT

Ischemic stroke is a clinical condition characterized by hypoperfusion of brain tissue, leading to oxygen and glucose deprivation, and the consequent neuronal loss. Numerous evidence suggests that the interaction between glial and neuronal cells exert beneficial effects after an ischemic event. Therefore, to explore potential protective mechanisms, it is important to develop models that allow studying neuron-glia interactions in an ischemic environment. Herein we present a simple approach to isolate astrocytes and neurons from the rat embryonic cortex, and that by using specific culture media, allows the establishment of neuron- or astrocyte-enriched cultures or neuron-glia cultures with high yield and reproducibility. To study the crosstalk between astrocytes and neurons, we propose an approach based on a co-culture system in which neurons cultured in coverslips are maintained in contact with a monolayer of astrocytes plated in multiwell plates. The two cultures are maintained apart by small paraffin spheres. This approach allows the independent manipulation and the application of specific treatments to each cell type, which represents an advantage in many studies. To simulate what occurs during an ischemic stroke, the cultures are subjected to an oxygen and glucose deprivation protocol. This protocol represents a useful tool to study the role of neuron-glia interactions in ischemic stroke.


Subject(s)
Cell Communication , Cell Culture Techniques/methods , Ischemia/pathology , Models, Biological , Neuroglia/pathology , Neurons/pathology , Animals , Cells, Cultured , Cerebral Cortex/embryology , Coculture Techniques , Embryo, Mammalian/cytology , Female , Glucose/metabolism , Oxygen/metabolism , Rats, Wistar , Reproducibility of Results
12.
Tissue Cell ; 66: 101400, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32933705

ABSTRACT

Although human brain represents only 2% of the body mass, it uses around 20 % of the organism energy. Due to the brain's limited energy storage, the oxygen and glucose necessary to support brain functions depends on the correct blood supply. The main components of the arteries are smooth muscle cells, which are considered the main regulators of vascular tone and blood flow distribution. The information currently available on the functioning of the cerebral arteries and their cell constituents is extremely scarce. Thus, the aim of this work was to develop an in vitro model of smooth muscle cells derived from rat middle cerebral artery. Explants were collected from rat middle cerebral artery and adhered to collagen-coated culture dishes. Immunocytochemical analysis showed that the cells present in the culture expressed α-actin, a protein characteristic of the contractile phenotype of these cells. In addition, these cells did not express the endothelial marker, vWF. To evaluate the functionality of these cells the response to contractile agents, serotonin and noradrenaline, and to relaxing agent, sodium nitroprusside was determine by Planar Cell Surface Area analysis. Together the data obtained show that the cell culture obtained through the procedure described resulted in cells presenting the markers characteristic of smooth muscle cells and maintaining the usual contractile response, indicating that the cells obtained through this may be used as a model for characterization and study of functional behavior of the middle cerebral artery, as well as interaction studies between vascular and neuronal system.


Subject(s)
Cell Separation , Cytoskeletal Proteins/metabolism , Middle Cerebral Artery/cytology , Muscle Proteins/metabolism , Myocytes, Smooth Muscle/cytology , Animals , Calcium-Binding Proteins/metabolism , Cells, Cultured , Microfilament Proteins/metabolism , Myocytes, Smooth Muscle/drug effects , Nitroprusside/pharmacology , Norepinephrine/pharmacology , Phenotype , Rats , Serotonin/pharmacology , Umbilical Cord/cytology , Calponins
13.
Transl Neurodegener ; 9: 11, 2020.
Article in English | MEDLINE | ID: mdl-32266064

ABSTRACT

Background: The identification of circulating biomarkers that closely correlate with Parkinson's Disease (PD) has failed several times in the past. Nevertheless, in this pilot study, a translational approach was conducted, allowing the evaluation of the plasma levels of two mitochondrial-related proteins, whose combination leads to a robust model with potential diagnostic value to discriminate the PD patients from matched controls. Methods: The proposed translational approach was initiated by the analysis of secretomes from cells cultured under control or well-defined oxidative stress conditions, followed by the identification of proteins related to PD pathologic mechanisms that were altered between the two states. This pipeline was further translated into the analysis of undepleted plasma samples from 28 control and 31 PD patients. Results: From the secretome analysis, several mitochondria-related proteins were found to be differentially released between control and stress conditions and to be able to distinguish the two secretomes. Similarly, two mitochondrial-related proteins were found to be significantly changed in a PD cohort compared to matched controls. Moreover, a linear discriminant model with potential diagnostic value to discriminate PD patients was obtained using the combination of these two proteins. Both proteins are associated with apoptotic mitochondrial changes, which may correspond to potential indicators of cell death. Moreover, one of these proteins, the VPS35 protein, was reported in plasma for the first time, and its quantification was only possible due to its previous identification in the secretome analysis. Conclusions: In this work, an adaptation of a translational pipeline for biomarker selection was presented and transposed to neurological diseases, in the present case Parkinson's Disease. The novelty and success of this pilot study may arise from the combination of: i) a translational research pipeline, where plasma samples are interrogated using knowledge previously obtained from the evaluation of cells' secretome under oxidative stress; ii) the combined used of statistical analysis and an informed selection of candidates based on their link with relevant disease mechanisms, and iii) the use of SWATH-MS, an untargeted MS method that allows a complete record of the analyzed samples and a targeted data extraction of the quantitative values of proteins previously identified.


Subject(s)
Mitochondrial Proteins/analysis , Parkinson Disease/diagnosis , Aged , Aged, 80 and over , Apoptosis , Biomarkers/analysis , Cells, Cultured , Cohort Studies , Female , HeLa Cells , Humans , Male , Middle Aged , Oxidative Stress , Pilot Projects , Proteomics , Translational Research, Biomedical , Vesicular Transport Proteins/blood
14.
Brain Res ; 1732: 146700, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32032613

ABSTRACT

The central nervous system (CNS) has a limited auto-regeneration capacity, which makes it challenging for the development of new therapies. Previous studies from our lab have demonstrated the applicability of human bone marrow mesenchymal stem cells (hBM-MSCs) secretome as a possible therapeutic tool for CNS. Astrocytes, glial cells present in all brain regions, are important players in brain function through their vast influence in extracellular homeostasis, neuro-vascular regulation, synaptic modulation and neurogenesis. Thus, in the present work, we aimed to evaluate the specific impact of MSCs secretome on hippocampal proliferation and astrocyte morphology, in both WT and dnSNARE mice, a transgenic model that presents impaired astrocytic exocytosis and consequently impaired astrocytic function. Results demonstrated increased levels of proliferation for WT when treated with secretome. Additionally, it was possible to observe that dnSNARE animals injected with hBM-MSCs secretome disclosed increased levels of proliferating GFAP stained cells at the SGZ. Morphometrical evaluation found increased process hypertrophy and branching of dnSNARE astrocytes when treated with secretome. These results are closely related with the trophic factors present in the secretome, namely FGF-2, BDNF, GDNF, IGF-1, VEGF, CADH2, PEDF and miR-16. Moreover, the impaired exocytosis of astrocytes may also have implications for the response to the proliferative stimulus, given the established autocrine signaling through this mechanism.


Subject(s)
Astrocytes/metabolism , Hippocampus/metabolism , Mesenchymal Stem Cells/metabolism , Signal Transduction/physiology , Astrocytes/cytology , Cell Proliferation/physiology , Cell Shape/physiology , Culture Media, Conditioned , Hippocampus/cytology , Humans
15.
Curr Neurovasc Res ; 16(5): 502-515, 2019.
Article in English | MEDLINE | ID: mdl-31738142

ABSTRACT

The neurovascular unit is a physiological unit present in the brain, which is constituted by elements of the nervous system (neurons and astrocytes) and the vascular system (endothelial and mural cells). This unit is responsible for the homeostasis and regulation of cerebral blood flow. There are two major types of mural cells in the brain, pericytes and smooth muscle cells. At the arterial level, smooth muscle cells are the main components that wrap around the outside of cerebral blood vessels and the major contributors to basal tone maintenance, blood pressure and blood flow distribution. They present several mechanisms by which they regulate both vasodilation and vasoconstriction of cerebral blood vessels and their regulation becomes even more important in situations of injury or pathology. In this review, we discuss the main regulatory mechanisms of brain smooth muscle cells and their contributions to the correct brain homeostasis.


Subject(s)
Arteries/physiology , Blood-Brain Barrier/physiology , Cerebrovascular Circulation/physiology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Animals , Brain/blood supply , Humans
17.
J Neurochem ; 149(1): 27-40, 2019 04.
Article in English | MEDLINE | ID: mdl-30570746

ABSTRACT

Selective activation of the G protein-coupled estrogen receptor has been proposed to avoid some of the side effects elicited by the activation of classical estrogen receptors α and ß. Although its contribution to neuroprotection triggered by estradiol in brain disorders has been explored, the results regarding ischemic stroke are contradictory, and currently, there is no consensus on the role that this receptor may play. The present study aimed to investigate the role of GPER in the ischemic insult. For that, primary cortical cultures exposed to oxygen and glucose deprivation (OGD) were used as a model. Our results demonstrate that neuronal survival was strongly affected by the ischemic insult and concurrent GPER activation with G1 had no further impact. In contrast, OGD had a smaller impact on astrocytes survival but G1, alone or combined with OGD, promoted their apoptosis. This effect was prevented by the GPER antagonist G15. The results also show that ischemia did not change the expression levels of GPER in neurons and astrocytes. In this study, we also demonstrate that selective activation of GPER induced astrocyte apoptosis via the phospholipase C pathway and subsequent intracellular calcium rise, whereas in neurons, this effect was not observed. Taken together, this evidence supports a direct impact of GPER activity on the viability of astrocytes, which seems to be associated with the regulation of different signaling pathways in astrocytes and neurons.


Subject(s)
Astrocytes/metabolism , Ischemia/metabolism , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Survival , Cells, Cultured , Cerebral Cortex/metabolism , Rats , Rats, Wistar , Signal Transduction/physiology
18.
Brain Behav Immun ; 64: 296-307, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28450223

ABSTRACT

Increasing evidence suggest that excessive inflammatory responses from overactivated microglia play a critical role in Parkinson's disease (PD), contributing to, or exacerbating, nigral dopaminergic (DA) degeneration. Recent results from our group and others demonstrated that selective activation of G protein-coupled estrogen receptor (GPER) with the agonist G1 can protect DA neurons from 1-methyl-4-phenylpyridinium (MPP+) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxins. However, it is not known whether modulation of microglial responses is one of the mechanisms by which G1 exerts its DA neuroprotective effects. We analyzed, in the N9 microglial cell line, the effect of G1 on microglial activation induced by lipopolysaccharide (LPS) exposure. The results revealed that G1 significantly decrease phagocytic activity, expression of inducible nitric oxide synthase (iNOS) and release of nitric oxide (NO) induced by LPS. To determine the relevance of this anti-inflammatory effect to the protection of nigral DA cells, the effect of G1 was analyzed in male mice injected unilaterally in the substantia nigra (SN) with LPS. Although G1 treatment did not decrease LPS-induced increase of ionized calcium binding adaptor molecule 1 (iba-1) positive cells it significantly reduced interleukin-1beta (IL-1ß), cluster of differentiation 68 (CD68) and iNOS mRNA levels, and totally inhibited nigral DA cell loss and, as a consequence, protected the motor function. In summary, our findings demonstrated that the G1 agonist is able to modulate microglial responses and to protect DA neurons and motor functions against a lesion induced by an inflammatory insult. Since G1 lacks the feminizing effects associated with agonists of the classical estrogen receptors (ERs), the use of G1 to selectively activate the GPER may be a promising strategy for the development of new therapeutics for the treatment of PD and other neuroinflammatory diseases.


Subject(s)
Dopaminergic Neurons/pathology , Encephalitis/metabolism , Microglia/physiology , Motor Activity , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Substantia Nigra/metabolism , Substantia Nigra/pathology , Animals , Cell Line , Dopaminergic Neurons/metabolism , Encephalitis/chemically induced , Encephalitis/complications , Lipopolysaccharides/administration & dosage , Male , Mice, Inbred C57BL , Microglia/metabolism , Parkinson Disease, Secondary/metabolism , Phagocytosis , Rats, Wistar , Receptors, G-Protein-Coupled/agonists , Tyrosine 3-Monooxygenase/metabolism
19.
Cell Mol Neurobiol ; 37(8): 1521-1528, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28315110

ABSTRACT

Cell cultures are characterized by their simplicity, controllability, and ability to provide detailed basic information on how a particular cell population responds to specific stimuli or insult. These characteristics led to their extensive application in the study of molecular interactions and represent a valuable tool in the study of different pathologies. However, due to the lack of interactions between the different components that form an in vivo system, the results obtained in pure cell cultures not always translate what occurs in vivo. In this context, the use of co-cultures has the advantage of allowing the study of interactions between different types of cells present in a tissue, which in many situations are determinant for the effects obtained. The present study aimed to characterize cortical neuron-glia and neuron-enriched primary cultures and evaluate their response to an ischemic insult. Cell viability was assessed by the MTT assay and cell number/phenotype was analyzed by immunocytochemistry in control cultures and in cells subjected to 4 h of oxygen and glucose deprivation. The results obtained demonstrate that astrocytes have a substantial impact on the injury induced by an ischemic insult, thus suggesting that the crosstalk between glia and neurons is crucial to the neuronal protection in conditions of ischemia.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/metabolism , Neuroglia/metabolism , Neurons/metabolism , Animals , Cell Hypoxia/physiology , Cells, Cultured , Cerebral Cortex/pathology , Coculture Techniques , Female , Neuroglia/pathology , Neurons/pathology , Pregnancy , Rats , Rats, Wistar
20.
Curr Pharm Des ; 23(5): 809-838, 2017.
Article in English | MEDLINE | ID: mdl-27928963

ABSTRACT

Parkinson's disease is an age-associated progressive neurodegenerative disorder that has gained crescent social and economic impact due to the aging of the western society. All current therapies are symptomatic and fail to reverse or halt the progression of dopaminergic neurons loss. The discovery of the capability of neurotrophic factors to protect these neurons lead numerous research groups to focus their efforts in developing therapies aiming at promoting the control of Parkinson´s disease through the delivery of neurotrophic factors to the brain or by boosting their endogenous levels. Both strategies were successful in inducing protection of dopaminergic neurons and motor recovery in preclinical models of the disease. Contrariwise, very limited success was obtained in clinical studies, where glial cell line-derived neurotrophic factor and neurturin were the neurotrophic factors of choice for Parkinson's disease therapy. These drawbacks motivate the development of novel forms of delivery or the modification of the injected molecules aiming at providing a more stable and effective administration with improved diffusion in the target tissue, and without the immune responses observed in the earliest clinical studies. Although promising results were obtained with some of these new approaches performed in experimental models of the disease, they were not yet tested in human studies. In this review, we present the current knowledge on neurotrophic factors and their role in Parkinson's disease, focusing on the strategies that have been developed to increase their levels in target areas of the brain to achieve protection of dopaminergic neurons and motor behaviour recovery.


Subject(s)
Nerve Growth Factors/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...