Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neurotoxicology ; 89: 55-66, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34999154

RESUMEN

Epidemiological studies reveal that air pollution exposure may exacerbate neurodegeneration. Ultrafine particles (UFPs) are pollutants that remain unregulated in ambient air by environmental agencies. Due to their small size (<100 nm), UFPs have the most potential to cross the bodily barriers and thus impact the brain. However, little information exists about how UFPs affect brain function. Alzheimer's disease (AD) is the most common form of dementia, which has been linked to air pollutant exposure, yet limited information is available on the mechanistic connection between them. This study aims to decipher the effects of UFPs in the brain and periphery using the 5xFAD mouse model of AD. In our study design, AD mice and their wildtype littermates were subjected to 2-weeks inhalation exposure of UFPs in a whole-body chamber. That subacute exposure did not affect the amyloid-beta accumulation. However, when multiple cytokines were analyzed, we found increased levels of proinflammatory cytokines in the brain and periphery, with a predominant alteration of interferon-gamma in response to UFP exposure in both genotypes. Following exposure, mitochondrial superoxide dismutase was significantly upregulated only in the 5xFAD hippocampi, depicting oxidative stress induction in the exposed AD mouse group. These data demonstrate that short-term exposure to inhaled UFPs induces inflammation without affecting amyloid-beta load. This study provides a better understanding of adverse effects caused by short-term UFP exposure in the brain and periphery, also in the context of AD.


Asunto(s)
Contaminantes Atmosféricos , Material Particulado , Contaminantes Atmosféricos/toxicidad , Péptidos beta-Amiloides , Animales , Inflamación/inducido químicamente , Exposición por Inhalación/efectos adversos , Exposición por Inhalación/análisis , Ratones , Tamaño de la Partícula , Material Particulado/toxicidad
2.
Int J Mol Sci ; 22(6)2021 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-33803024

RESUMEN

Microglia are resident immune cells of the central nervous system and play critical roles during the development, homeostasis, and pathologies of the brain. Originated from yolk sac erythromyeloid progenitors, microglia immigrate into the embryonic brain parenchyma to undergo final postnatal differentiation and maturation driven by distinct chemokines, cytokines, and growth factors. Among them, TGFß1 is an important regulator of microglial functions, mediating homeostasis, anti-inflammation, and triggering the expression of microglial homeostatic signature genes. Since microglia studies are mainly based on rodent cells and the isolation of homeostatic microglia from human tissue is challenging, human-induced pluripotent stem cells have been successfully differentiated into microglia-like cells recently. However, employed differentiation protocols strongly vary regarding used cytokines and growth factors, culture conditions, time span, and cell yield. Moreover, the incomplete differentiation of human microglia can hamper the similarity to primary human microglia and dramatically influence the outcome of follow-up studies with these differentiated cells. This review summarizes the current knowledge of the molecular mechanisms driving rodent microglia differentiation in vivo, further compares published differentiation protocols, and highlights the potential of TGFß as an essential maturation factor.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Microglía/citología , Animales , Uniones Célula-Matriz/metabolismo , Humanos , Microglía/metabolismo , Modelos Biológicos , Factor de Crecimiento Transformador beta/metabolismo
3.
Cells ; 11(1)2021 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-35011667

RESUMEN

Human pluripotent stem cell (hPSC)-derived neuron cultures have emerged as models of electrical activity in the human brain. Microelectrode arrays (MEAs) measure changes in the extracellular electric potential of cell cultures or tissues and enable the recording of neuronal network activity. MEAs have been applied to both human subjects and hPSC-derived brain models. Here, we review the literature on the functional characterization of hPSC-derived two- and three-dimensional brain models with MEAs and examine their network function in physiological and pathological contexts. We also summarize MEA results from the human brain and compare them to the literature on MEA recordings of hPSC-derived brain models. MEA recordings have shown network activity in two-dimensional hPSC-derived brain models that is comparable to the human brain and revealed pathology-associated changes in disease models. Three-dimensional hPSC-derived models such as brain organoids possess a more relevant microenvironment, tissue architecture and potential for modeling the network activity with more complexity than two-dimensional models. hPSC-derived brain models recapitulate many aspects of network function in the human brain and provide valid disease models, but certain advancements in differentiation methods, bioengineering and available MEA technology are needed for these approaches to reach their full potential.


Asunto(s)
Encéfalo/fisiología , Modelos Biológicos , Células Madre Pluripotentes/metabolismo , Humanos , Microelectrodos , Neuronas/fisiología , Organoides/fisiología
4.
Cells ; 11(1)2021 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-35011686

RESUMEN

Human cerebral organoids, derived from induced pluripotent stem cells, offer a unique in vitro research window to the development of the cerebral cortex. However, a key player in the developing brain, the microglia, do not natively emerge in cerebral organoids. Here we show that erythromyeloid progenitors (EMPs), differentiated from induced pluripotent stem cells, migrate to cerebral organoids, and mature into microglia-like cells and interact with synaptic material. Patch-clamp electrophysiological recordings show that the microglia-like population supported the emergence of more mature and diversified neuronal phenotypes displaying repetitive firing of action potentials, low-threshold spikes and synaptic activity, while multielectrode array recordings revealed spontaneous bursting activity and increased power of gamma-band oscillations upon pharmacological challenge with NMDA. To conclude, microglia-like cells within the organoids promote neuronal and network maturation and recapitulate some aspects of microglia-neuron co-development in vivo, indicating that cerebral organoids could be a useful biorealistic human in vitro platform for studying microglia-neuron interactions.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Microglía/metabolismo , Neurogénesis/genética , Neuronas/metabolismo , Organoides/metabolismo , Adolescente , Adulto , Anciano , Diferenciación Celular , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
5.
Neurochem Int ; 136: 104715, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32169588

RESUMEN

Every second we inhale a danger in the air; many particles in the atmosphere can influence our lives. Outdoor air pollution, especially particulate matter is the largest environmental risk factor and has been associated with many cardiovascular and lung diseases. Importantly, air pollution has recently been discovered to also impact the brain. Here, we review the effects of air pollution on glial cells of the brain, astrocytes and microglia, and the tightly controlled interplay between these cell types. We focus on how traffic related air pollutants which include both gaseous and particulate emissions and their secondary products influence the intercellular communication of microglia and astrocytes. Finally, we place these air pollution and glial interactions in a larger context by discussing their impact on neurodegeneration.


Asunto(s)
Contaminantes Atmosféricos/efectos adversos , Contaminación del Aire/efectos adversos , Astrocitos/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Contaminantes Atmosféricos/análisis , Contaminación del Aire/análisis , Animales , Astrocitos/efectos de los fármacos , Exposición a Riesgos Ambientales/efectos adversos , Humanos , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo
6.
Stem Cell Reports ; 13(4): 669-683, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31522977

RESUMEN

Here we elucidate the effect of Alzheimer disease (AD)-predisposing genetic backgrounds, APOE4, PSEN1ΔE9, and APPswe, on functionality of human microglia-like cells (iMGLs). We present a physiologically relevant high-yield protocol for producing iMGLs from induced pluripotent stem cells. Differentiation is directed with small molecules through primitive erythromyeloid progenitors to re-create microglial ontogeny from yolk sac. The iMGLs express microglial signature genes and respond to ADP with intracellular Ca2+ release distinguishing them from macrophages. Using 16 iPSC lines from healthy donors, AD patients and isogenic controls, we reveal that the APOE4 genotype has a profound impact on several aspects of microglial functionality, whereas PSEN1ΔE9 and APPswe mutations trigger minor alterations. The APOE4 genotype impairs phagocytosis, migration, and metabolic activity of iMGLs but exacerbates their cytokine secretion. This indicates that APOE4 iMGLs are fundamentally unable to mount normal microglial functionality in AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Apolipoproteína E4/genética , Diferenciación Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Microglía/metabolismo , Fenotipo , Presenilina-1/genética , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Apolipoproteína E4/metabolismo , Calcio/metabolismo , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , Hematopoyesis , Humanos , Células Madre Pluripotentes Inducidas/citología , Mediadores de Inflamación/metabolismo , Microglía/citología , Mutación , Fagocitosis , Presenilina-1/metabolismo , Proteolisis
7.
Glia ; 67(1): 146-159, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30453390

RESUMEN

Astrocytes are the gatekeepers of neuronal energy supply. In neurodegenerative diseases, bioenergetics demand increases and becomes reliant upon fatty acid oxidation as a source of energy. Defective fatty acid oxidation and mitochondrial dysfunctions correlate with hippocampal neurodegeneration and memory deficits in Alzheimer's disease (AD), but it is unclear whether energy metabolism can be targeted to prevent or treat the disease. Here we show for the first time an impairment in fatty acid oxidation in human astrocytes derived from induced pluripotent stem cells of AD patients. The impairment was corrected by treatment with a synthetic peroxisome proliferator activated receptor delta (PPARß/δ) agonist GW0742 which acts to regulate an array of genes governing cellular metabolism. GW0742 enhanced the expression of CPT1a, the gene encoding for a rate-limiting enzyme of fatty acid oxidation. Similarly, treatment of a mouse model of AD, the APP/PS1-mice, with GW0742 increased the expression of Cpt1a and concomitantly reversed memory deficits in a fear conditioning test. Although the GW0742-treated mice did not show altered astrocytic glial fibrillary acidic protein-immunoreactivity or reduction in amyloid beta (Aß) load, GW0742 treatment increased hippocampal neurogenesis and enhanced neuronal differentiation of neuronal progenitor cells. Furthermore, GW0742 prevented Aß-induced impairment of long-term potentiation in hippocampal slices. Collectively, these data suggest that PPARß/δ-agonism alleviates AD related deficits through increasing fatty acid oxidation in astrocytes and improves cognition in a transgenic mouse model of AD.


Asunto(s)
Astrocitos/metabolismo , Ácidos Grasos/metabolismo , PPAR delta/metabolismo , PPAR-beta/metabolismo , Presenilina-1/metabolismo , Tiazoles/farmacología , Adulto , Animales , Astrocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Exones/efectos de los fármacos , Exones/fisiología , Femenino , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Oxidación-Reducción/efectos de los fármacos , PPAR delta/agonistas , PPAR-beta/agonistas , Distribución Aleatoria
8.
Dis Model Mech ; 10(9): 1089-1100, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28733362

RESUMEN

Neural stem/progenitor cells (NPCs) generate new neurons in the brain throughout an individual's lifetime in an intricate process called neurogenesis. Neurogenic alterations are a common feature of several adult-onset neurodegenerative diseases. The neuronal ceroid lipofuscinoses (NCLs) are the most common group of inherited neurodegenerative diseases that mainly affect children. Pathological features of the NCLs include accumulation of lysosomal storage material, neuroinflammation and neuronal degeneration, yet the exact cause of this group of diseases remains poorly understood. The function of the CLN5 protein, causative of the CLN5 disease form of NCL, is unknown. In the present study, we sought to examine neurogenesis in the neurodegenerative disorder caused by loss of Cln5 Our findings demonstrate a newly identified crucial role for CLN5 in neurogenesis. We report for the first time that neurogenesis is increased in Cln5-deficient mice, which model the childhood neurodegenerative disorder caused by loss of Cln5 Our results demonstrate that, in Cln5 deficiency, proliferation of NPCs is increased, NPC migration is reduced and NPC differentiation towards the neuronal lineage is increased concomitantly with functional alterations in the NPCs. Moreover, the observed impairment in neurogenesis is correlated with increased expression of the pro-inflammatory cytokine IL-1ß. A full understanding of the pathological mechanisms that lead to disease and the function of the NCL proteins are critical for designing effective therapeutic approaches for this devastating neurodegenerative disorder.


Asunto(s)
Hipocampo/metabolismo , Hipocampo/patología , Glicoproteínas de Membrana/deficiencia , Neurogénesis , Lipofuscinosis Ceroideas Neuronales/metabolismo , Lipofuscinosis Ceroideas Neuronales/patología , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Niño , Modelos Animales de Enfermedad , Humanos , Interleucina-1beta/farmacología , Proteínas de Membrana de los Lisosomas , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neurogénesis/efectos de los fármacos , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo
9.
Front Cell Neurosci ; 10: 279, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27994540

RESUMEN

Background: Accumulation of amyloid ß (Aß) is one of the main hallmarks of Alzheimer's disease (AD). The enhancement of Aß clearance may provide therapeutic means to restrict AD pathology. The cellular responses to different forms of Aß in monocytic cells are poorly known. We aimed to study whether different forms of Aß induce inflammatory responses in monocytic phagocytes and how Aß may affect monocytic cell survival and function to retain phagocytosis in Aß-laden environment. Methods: Monocytic cells were differentiated from bone marrow hematopoietic stem cells (HSC) in the presence of macrophage-colony stimulating factor. Monocytic cells were stimulated with synthetic Aß42 and intracellular calcium responses were recorded with calcium imaging. The formation of reactive oxygen species (ROS), secretion of cytokines and cell viability were also assessed. Finally, monocytic cells were introduced to native Aß deposits ex vivo and the cellular responses in terms of cell viability, pro-inflammatory activation and phagocytosis were determined. The ability of monocytic cells to phagocytose Aß plaques was determined after intrahippocampal transplantation in vivo. Results: Freshly solubilized Aß induced calcium oscillations, which persisted after removal of the stimulus. After few hours of aggregation, Aß was not able to induce oscillations in monocytic cells. Instead, lipopolysaccharide (LPS) induced calcium responses divergent from Aß-induced response. Furthermore, while LPS induced massive production of pro-inflammatory cytokines, neither synthetic Aß species nor native Aß deposits were able to induce pro-inflammatory activation of monocytic cells, contrary to primary microglia. Finally, monocytic cells retained their viability in the presence of Aß and exhibited phagocytic activity towards native fibrillar Aß deposits and congophilic Aß plaques. Conclusion: Monocytic cells carry diverse cellular responses to Aß and inflammatory stimulus LPS. Even though Aß species cause specific responses in calcium signaling, they completely lack the ability to induce pro-inflammatory phenotype of monocytic cells. Monocytes retain their viability and function in Aß-laden brain.

10.
Aging Dis ; 7(4): 450-65, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27493832

RESUMEN

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

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...