Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
BMC Neurosci ; 25(1): 29, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926677

RESUMEN

BACKGROUND: Astrocytes are the most abundant cell type of the central nervous system and are fundamentally involved in homeostasis, neuroprotection, and synaptic plasticity. This regulatory function of astrocytes on their neighboring cells in the healthy brain is subject of current research. In the ischemic brain we assume disease specific differences in astrocytic acting. The renin-angiotensin-aldosterone system regulates arterial blood pressure through endothelial cells and perivascular musculature. Moreover, astrocytes express angiotensin II type 1 and 2 receptors. However, their role in astrocytic function has not yet been fully elucidated. We hypothesized that the angiotensin II receptors impact astrocyte function as revealed in an in vitro system mimicking cerebral ischemia. Astrocytes derived from neonatal wistar rats were exposed to telmisartan (angiotensin II type 1 receptor-blocker) or PD123319 (angiotensin II type 2 receptor-blocker) under normal conditions (control) or deprivation from oxygen and glucose. Conditioned medium (CM) of astrocytes was harvested to elucidate astrocyte-mediated indirect effects on microglia and cortical neurons. RESULT: The blockade of angiotensin II type 1 receptor by telmisartan increased the survival of astrocytes during ischemic conditions in vitro without affecting their proliferation rate or disturbing their expression of S100A10, a marker of activation. The inhibition of the angiotensin II type 2 receptor pathway by PD123319 resulted in both increased expression of S100A10 and proliferation rate. The CM of telmisartan-treated astrocytes reduced the expression of pro-inflammatory mediators with simultaneous increase of anti-inflammatory markers in microglia. Increased neuronal activity was observed after treatment of neurons with CM of telmisartan- as well as PD123319-stimulated astrocytes. CONCLUSION: Data show that angiotensin II receptors have functional relevance for astrocytes that differs in healthy and ischemic conditions and effects surrounding microglia and neuronal activity via secretory signals. Above that, this work emphasizes the strong interference of the different cells in the CNS and that targeting astrocytes might serve as a therapeutic strategy to influence the acting of glia-neuronal network in de- and regenerative context.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II , Bloqueadores del Receptor Tipo 2 de Angiotensina II , Astrocitos , Accidente Cerebrovascular Isquémico , Microglía , Neuronas , Ratas Wistar , Receptor de Angiotensina Tipo 1 , Receptor de Angiotensina Tipo 2 , Telmisartán , Animales , Ratas , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 2 de Angiotensina II/farmacología , Animales Recién Nacidos , Astrocitos/metabolismo , Astrocitos/efectos de los fármacos , Bencimidazoles/farmacología , Comunicación Celular/fisiología , Comunicación Celular/efectos de los fármacos , Células Cultivadas , Imidazoles/farmacología , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Microglía/metabolismo , Microglía/efectos de los fármacos , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Piridinas/farmacología , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Telmisartán/farmacología
2.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34429357

RESUMEN

The development of the cerebral cortex relies on the controlled division of neural stem and progenitor cells. The requirement for precise spatiotemporal control of proliferation and cell fate places a high demand on the cell division machinery, and defective cell division can cause microcephaly and other brain malformations. Cell-extrinsic and -intrinsic factors govern the capacity of cortical progenitors to produce large numbers of neurons and glia within a short developmental time window. In particular, ion channels shape the intrinsic biophysical properties of precursor cells and neurons and control their membrane potential throughout the cell cycle. We found that hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channel subunits are expressed in mouse, rat, and human neural progenitors. Loss of HCN channel function in rat neural stem cells impaired their proliferation by affecting the cell-cycle progression, causing G1 accumulation and dysregulation of genes associated with human microcephaly. Transgene-mediated, dominant-negative loss of HCN channel function in the embryonic mouse telencephalon resulted in pronounced microcephaly. Together, our findings suggest a role for HCN channel subunits as a part of a general mechanism influencing cortical development in mammals.


Asunto(s)
Proliferación Celular/fisiología , Corteza Cerebral/embriología , Canalopatías/etiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Microcefalia/etiología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Animales , Ciclo Celular , Muerte Celular , Células Cultivadas , Corteza Cerebral/citología , Canalopatías/embriología , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/fisiología , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/antagonistas & inhibidores , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Ratones , Ratones Transgénicos , Microcefalia/embriología , Células-Madre Neurales/metabolismo , Ratas
3.
Eur J Neurol ; 30(12): 3979-3981, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37584071

RESUMEN

Although-considering the risk-benefit ratio-botulinum neurotoxin A (BoNT/A) is unequivocally recommended to treat severe neurological diseases such as dystonia, this has not yet been determined for its endoscopic intragastric injection aimed at weight reduction in obesity. However, severe adverse effects of intragastric BoNT/A had not yet been reported, prompting some European countries to endorse its (off-label) use and treat patients transnationally. We here present three cases of botulism after intragastric BoNT/A injections for obesity treatment in a Turkish hospital. Patients presented with cranial nerve affection, bulbar symptoms, and descending paresis, and benefited from treatment with BoNT antitoxin and pyridostigmine. We assume that iatrogenic botulism was induced by overdosing in combination with toxin spread via the highly vascularized gastric tissue. Of note, within a few weeks, more than 80 cases of iatrogenic botulism were reported across Europe after identical intragastric BoNT/A injections. These cases demonstrate the risks of BoNT/A injections if they are not applied within the limits of evidence-based medicine. There is a need for international guidelines to define the indication and a safe dosing scheme, especially in the context of medical tourism.


Asunto(s)
Toxinas Botulínicas Tipo A , Botulismo , Humanos , Botulismo/etiología , Botulismo/inducido químicamente , Toxinas Botulínicas Tipo A/efectos adversos , Enfermedad Iatrogénica , Pérdida de Peso , Obesidad
4.
J Neurosci Res ; 99(11): 2822-2843, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34510519

RESUMEN

The glycoprotein osteopontin is highly upregulated in central nervous system (CNS) disorders such as ischemic stroke. Osteopontin regulates cell growth, cell adhesion, homeostasis, migration, and survival of various cell types. Accordingly, osteopontin is considered an essential regulator of regeneration and repair in the ischemic milieu. Astrocytes are the most abundant cells in the CNS and play significant roles in health and disease. Astrocytes are involved in homeostasis, promote neuroprotection, and regulate synaptic plasticity. Upon activation, astrocytes may adopt different phenotypes, termed A1 and A2. The direct effects of osteopontin on astrocytes, especially in distinct activation states, are yet unknown. The current study aimed to elucidate the impact of osteopontin on resting and active astrocytes. We established an inflammatory in vitro model of activated (A1) primary astrocytes derived from neonatal wistar rats by exposure to a distinct combination of proinflammatory cytokines. To model ischemic stroke in vitro, astrocytes were subjected to oxygen and glucose deprivation (OGD) in the presence or absence of osteopontin. Osteopontin modulated the activation phenotype by attenuating A1- and restoring A2-marker expression without compromising the active astrocytes' immunocompetence. Osteopontin promoted the proliferation of active and the migration of resting astrocytes. Following transient OGD, osteopontin mitigated the delayed ongoing death of primary astrocytes, promoting their survival. Data suggest that osteopontin differentially regulates essential functions of resting and active astrocytes and confirm a significant regulatory role of osteopontin in an in vitro ischemia model. Furthermore, the data suggest that osteopontin constitutes a promising target for experimental therapies modulating neuroregeneration and repair.


Asunto(s)
Astrocitos , Osteopontina , Animales , Astrocitos/metabolismo , Proliferación Celular , Plasticidad Neuronal , Fenotipo , Ratas
5.
J Neuroinflammation ; 17(1): 33, 2020 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-31980036

RESUMEN

BACKGROUND: In cerebral ischemia, microglia have a dichotomous role in keeping the balance between pro- and anti-inflammatory mediators to avoid deleterious chronic inflammation and to leverage repair processes. METHODS: We examined functional and inflammatory markers in primary rat microglia in vitro after oxygen-glucose deprivation (OGD) or glucose deprivation (aglycemia). We then investigated the preconditioning effect of OGD or aglycemia upon a subsequent strong inflammatory stimulus, here lipopolysaccharides (LPS). Moreover, an "in vitro brain model" of neurons and glia, differentiated from primary rat neural stem cells, was exposed to OGD or aglycemia. Conditioned medium (CM) of this neuronal/glial co-culture was then used to condition microglia, followed by LPS as a "second hit." RESULTS: OGD or aglycemia at sublethal doses did not significantly affect microglia function, including the expression of inflammatory markers. However, preconditioning with either OGD or aglycemia led to a decreased pro-inflammatory response to a subsequent stimulus with LPS. Interestingly, the anti-inflammatory markers IGF-1 and IL-10 were additionally reduced after such preconditioning, while expression of CD206 remained unaffected. Treatment with CM from the neuronal/glial co-culture alone did not affect the expression of inflammatory markers in microglia. In contrast, treatment with CM increased the expression of both pro- and anti-inflammatory markers in microglia upon a second hit with LPS. Interestingly, this effect could be attenuated in microglia treated with CM from neuronal/glia co-cultures preconditioned with OGD or aglycemia. CONCLUSIONS: Data suggest specific and distinct microglia signatures in response to metabolic stress. While metabolic stress directly and indirectly applied to microglia did not mitigate their subsequent response to inflammation, preconditioning with metabolic stress factors such as OGD and aglycemia elicited a decreased inflammatory response to a subsequent inflammation stimulus.


Asunto(s)
Inflamación/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Receptor Cross-Talk/fisiología , Estrés Fisiológico/fisiología , Animales , Isquemia Encefálica/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Glucosa/deficiencia , Inflamación/inducido químicamente , Lipopolisacáridos/farmacología , Ratas
6.
J Neuroinflammation ; 17(1): 100, 2020 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-32248813

RESUMEN

BACKGROUND: Microglia are essential to maintain cell homeostasis in the healthy brain and are activated after brain injury. Upon activation, microglia polarize towards different phenotypes. The course of microglia activation is complex and depends on signals in the surrounding milieu. Recently, it has been suggested that microglia respond to ion currents, as a way of regulating their activity and function. METHODS AND RESULTS: Under the hypothesis that HCN and KCNQ/Kv7 channels impact on microglia, we studied primary rat microglia in the presence or absence of specific pharmacological blockade or RNA silencing. Primary microglia expressed the subunits HCN1-4, Kv7.2, Kv7.3, and Kv7.5. The expression of HCN2, as well as Kv7.2 and Kv7.3, varied among different microglia phenotypes. The pharmacological blockade of HCN channels by ZD7288 resulted in cell depolarization with slowly rising intracellular calcium levels, leading to enhanced survival and reduced proliferation rates of resting microglia. Furthermore, ZD7288 treatment, as well as knockdown of HCN2 RNA by small interfering RNA, resulted in an attenuation of later microglia activation-both towards the anti- and pro-inflammatory phenotype. However, HCN channel inhibition enhanced the phagocytic capacity of IL4-stimulated microglia. Blockade of Kv7/KCNQ channel by XE-991 exclusively inhibited the migratory capacity of resting microglia. CONCLUSION: These observations suggest that the HCN current contributes to various microglia functions and impacts on the course of microglia activation, while the Kv7/KCNQ channels affect microglia migration. Characterizing the role of HCN channels in microglial functioning may offer new therapeutic approaches for targeted modulation of neuroinflammation as a hallmark of various neurological disorders.


Asunto(s)
Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Microglía/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/metabolismo , Animales , Calcio/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/antagonistas & inhibidores , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Microglía/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Canales de Potasio con Entrada de Voltaje/genética , Pirimidinas/farmacología , Interferencia de ARN , Ratas , Ratas Wistar
7.
J Neuroinflammation ; 15(1): 226, 2018 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-30103769

RESUMEN

BACKGROUND: Microglia-the resident immune cells of the brain-are activated after brain lesions, e.g., cerebral ischemia, and polarize towards a classic "M1" pro-inflammatory or an alternative "M2" anti-inflammatory phenotype following characteristic temporo-spatial patterns, contributing either to secondary tissue damage or to regenerative responses. They closely interact with endogenous neural stem cells (NSCs) residing in distinct niches of the adult brain. The current study aimed at elucidating the dynamics of microglia polarization and their differential effects on NSC function. RESULTS: Primary rat microglia in vitro were polarized towards a M1 phenotype by LPS, or to a M2 phenotype by IL4, while simultaneous exposure to LPS plus IL4 resulted in a hybrid phenotype expressing both M1- and M2-characteristic markers. M2 microglia migrated less but exhibit higher phagocytic activity than M1 microglia. Defined mediators switched microglia from one polarization state to the other, a process more effective when transforming M2 microglia towards M1 than vice versa. Polarized microglia had differential effects on the differentiation potential of NSCs in vitro and in vivo, with M1 microglia promoting astrocytogenesis, while M2 microglia supported neurogenesis. Regardless of their polarization, microglia inhibited NSC proliferation, increased NSC migration, and accelerated NSC differentiation. CONCLUSION: Overall, this study shed light on the complex conditions governing microglia polarization and the effects of differentially polarized microglia on critical functions of NSCs in vitro and in vivo. Refining the understanding of microglia activation and their modulatory effects on NSCs is likely to facilitate the development of innovative therapeutic concepts supporting the innate regenerative capacity of the brain.


Asunto(s)
Microglía/fisiología , Células-Madre Neurales/fisiología , Animales , Animales Recién Nacidos , Diferenciación Celular/fisiología , Movimiento Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Polaridad Celular/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Interleucina-4/farmacología , Interleucina-6/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Lipopolisacáridos/farmacología , Masculino , Microglía/efectos de los fármacos , Microglía/ultraestructura , Células-Madre Neurales/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fagocitosis/fisiología , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/metabolismo
8.
Mediators Inflamm ; 2017: 7189421, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29104378

RESUMEN

BACKGROUND: Focal cerebral ischemia induces distinct neuroinflammatory processes. We recently reported the extracellular phosphor-glyco-protein osteopontin (OPN) to directly affect primary microglia in vitro, promoting survival while shifting their inflammatory profile towards a more neutral phenotype. We here assessed the effects of OPN on microglia after stroke in vivo, with focus on infarct demarcation. METHODS: Animals underwent focal photothrombotic stroke and were injected intracerebroventricularly with 500 µg OPN or vehicle. Immunohistochemistry assessed neuronal damage and infarct volume, neovascularisation, glial scar formation, microglial activation, and M1 and M2 polarisation. RESULTS: After photothrombotic stroke, areas covered by M1 and M2 microglia substantially overlapped. OPN treatment reduced that overlap, with microglia appearing more spread out and additionally covering the infarct core. OPN additionally modulated the quantity of microglia subpopulations, reducing iNOS+ M1 cells while increasing M2 microglia, shifting the M1/M2 balance towards an M2 phenotype. Moreover, OPN polarized astrocytes towards the infarct. CONCLUSION: Microglial activation and M1 and M2 polarization have distinct but overlapping spatial patterns in permanent focal ischemia. Data suggest that OPN is involved in separating M1 and M2 subpopulations, as well as in shifting microglia polarization towards the M2 phenotype modulating beneficially inflammatory responses after focal infarction.


Asunto(s)
Microglía/efectos de los fármacos , Osteopontina/uso terapéutico , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Inmunohistoquímica , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Microglía/metabolismo , Ratas , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo
9.
J Neurosci Res ; 94(2): 149-60, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26525774

RESUMEN

Mobilizing endogenous neural stem cells (NSCs) in the adult brain is designed to enhance the brain's regenerative capacity after cerebral lesions, e.g., as a result of stroke. Cerebral ischemia elicits neuroinflammatory processes affecting NSCs in multiple ways, the precise mechanisms of which currently remain elusive. An inhibitory effect of minocycline on microglia activation, a hallmark of postischemic neuroinflammation, has already been demonstrated in clinical trials, showing minocycline to be safe and potentially effective in ischemic stroke. Here we investigate the direct effects of minocycline and of proinflammatory cytokines on the differentiation potential of NSCs in vitro and in vivo. Primary fetal rat NSCs were treated with minocycline plus a combination of the proinflammatory cytokines tumor necrosis factor-α, interleukin 1ß, and interleukin 6. The differentiation fate of NSCs was assessed immunocytochemically. To investigate the effects of minocycline and inflammation in vivo, minocycline or lipopolysaccharides were injected intraperitoneally into adult rats, with subsequent immunohistochemistry. Minocycline alone did not affect the differentiation potential of NSCs in vivo or in vitro. In contrast, proinflammatory cytokines accelerated the differentiation of NSCs, promoting an astrocytic fate while inhibiting neurogenesis in vitro and in vivo. It is interesting to note that minocycline counteracted this cytokine-induced rapid astrocytic differentiation and restored the neurogenic and oligodendrogliogenic potential of NSCs. Data suggest that minocycline antagonizes the rapid glial differentiation induced by proinflammatory cytokines following cerebral ischemia but without having a direct effect on the differentiation potential of NSCs. Thus, minocycline constitutes a promising drug for stroke research, counteracting the detrimental effects of postischemic neuroinflammation in multiple ways.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Citocinas/farmacología , Minociclina/farmacología , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , 2',3'-Nucleótido Cíclico Fosfodiesterasas/metabolismo , Animales , Antígenos/metabolismo , Astrocitos/efectos de los fármacos , Células Cultivadas , Combinación de Medicamentos , Embrión de Mamíferos , Proteína Ácida Fibrilar de la Glía/metabolismo , Lipopolisacáridos/farmacología , Masculino , Proteínas del Tejido Nervioso/metabolismo , Proteoglicanos/metabolismo , Ratas , Ratas Wistar , Factores de Transcripción SOXB1/metabolismo , Factores de Tiempo , Tubulina (Proteína)/metabolismo
10.
NMR Biomed ; 28(2): 231-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25521600

RESUMEN

Transcranial direct current stimulation (tDCS) is used in numerous clinical studies and considered an effective and versatile add-on therapy in neurorehabilitation. To date, however, the underlying neurobiological mechanisms remain elusive. In a rat model of tDCS, we recently observed a polarity-dependent accumulation of endogenous neural stem cells (NSCs) in the stimulated cortex. Based upon these findings, we hypothesized that tDCS may exert a direct migratory effect on endogenous NSCs towards the stimulated cortex. Using noninvasive imaging, we here investigated whether tDCS may also cause a directed migration of engrafted NSCs. Murine NSCs were labeled with superparamagnetic particles of iron oxide (SPIOs) and implanted into rat striatum and corpus callosum. MRI was performed (i) immediately after implantation and (ii) after 10 tDCS sessions of anodal or cathodal polarity. Sham-stimulated rats served as control. Imaging results were validated ex vivo using immunohistochemistry. Overall migratory activity of NSCs almost doubled after anodal tDCS. However, no directed migration within the electric field (i.e. towards or away from the electrode) could be observed. Rather, an undirected outward migration from the center of the graft was detected. Xenograft transplantation induced a neuroinflammatory response that was significantly enhanced following cathodal tDCS. This inflammatory response did not impact negatively on the survival of implanted NSCs. Data suggest that anodal tDCS increases the undirected migratory activity of implanted NSCs. Since the electric field did not guide implanted NSCs over large distances, previously observed polarity-dependent accumulation of endogenous NSCs in the cortex might have originated from local proliferation. Results enhance our understanding of the neurobiological mechanisms underlying tDCS, and may thereby help to develop a targeted and sustainable application of tDCS in clinical practice.


Asunto(s)
Encéfalo/metabolismo , Movimiento Celular , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Estimulación Transcraneal de Corriente Directa , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Electrodos , Inmunidad/efectos de los fármacos , Inmunohistoquímica , Hierro/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Imagen por Resonancia Magnética , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Células-Madre Neurales/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Ratas Wistar
11.
Front Pharmacol ; 13: 1038285, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36408236

RESUMEN

Glia are critical players in defining synaptic contacts and maintaining neuronal homeostasis. Both astrocytes as glia of the central nervous system (CNS), as well as satellite glial cells (SGC) as glia of the peripheral nervous system (PNS), intimately interact with microglia, especially under pathological conditions when glia regulate degenerative as well as regenerative processes. The chemotherapeutic agent paclitaxel evokes peripheral neuropathy and cognitive deficits; however, the mechanisms underlying these diverse clinical side effects are unclear. We aimed to elucidate the direct effects of paclitaxel on the function of astrocytes, microglia, and SGCs, and their glia-glia and neuronal-glia interactions. After intravenous application, paclitaxel was present in the dorsal root ganglia of the PNS and the CNS of rodents. In vitro, SGC enhanced the expression of pro-inflammatory factors and reduced the expression of neurotrophic factor NT-3 upon exposure to paclitaxel, resulting in predominantly neurotoxic effects. Likewise, paclitaxel induced a switch towards a pro-inflammatory phenotype in microglia, exerting neurotoxicity. In contrast, astrocytes expressed neuroprotective markers and increasingly expressed S100A10 after paclitaxel exposure. Astrocytes, and to a lesser extent SGCs, had regulatory effects on microglia independent of paclitaxel exposure. Data suggest that paclitaxel differentially modulates glia cells regarding their (neuro-) inflammatory and (neuro-) regenerative properties and also affects their interaction. By elucidating those processes, our data contribute to the understanding of the mechanistic pathways of paclitaxel-induced side effects in CNS and PNS.

12.
Neurorehabil Neural Repair ; 36(10-11): 701-714, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36124996

RESUMEN

BACKGROUND: Transcranial direct current stimulation (tDCS) promotes recovery after stroke in humans. The underlying mechanisms, however, remain to be elucidated. Animal models suggest tDCS effects on neuroinflammation, stem cell proliferation, neurogenesis, and neural plasticity. OBJECTIVE: In a longitudinal study, we employed tDCS in the subacute and chronic phase after experimental focal cerebral ischemia in mice to explore the relationship between functional recovery and cellular processes. METHODS: Mice received photothrombosis in the right motor cortex, verified by Magnetic Resonance Imaging. A composite neuroscore quantified subsequent functional deficits. Mice received tDCS daily: either 5 sessions from day 5 to 9, or 10 sessions with days 12 to 16 in addition. TDCS with anodal or cathodal polarity was compared to sham stimulation. Further imaging to assess proliferation and neuroinflammation was performed by immunohistochemistry at different time points and Positron Emission Tomography at the end of the observation time of 3 weeks. RESULTS: Cathodal tDCS at 198 kC/m2 (220 A/m2) between days 5 and 9 accelerated functional recovery, increased neurogenesis, decreased microglial activation, and mitigated CD16/32-expression associated with M1-phenotype. Anodal tDCS exerted similar effects on neurogenesis and microglial polarization but not on recovery of function or microglial activation. TDCS on days 12 to 16 after stroke did not induce any further effects, suggesting that the therapeutic time window was closed by then. CONCLUSION: Overall, data suggest that non-invasive neuromodulation by tDCS impacts neurogenesis and microglial activation as critical cellular processes influencing functional recovery during the early phase of regeneration from focal cerebral ischemia.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular , Estimulación Transcraneal de Corriente Directa , Humanos , Animales , Ratones , Estimulación Transcraneal de Corriente Directa/métodos , Recuperación de la Función , Estudios Longitudinales , Isquemia Encefálica/diagnóstico por imagen , Isquemia Encefálica/terapia , Isquemia Encefálica/complicaciones , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/terapia , Infarto Cerebral/complicaciones
13.
J Neurosci ; 30(18): 6454-60, 2010 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-20445071

RESUMEN

Neural stem cells reside in two major niches in the adult brain [i.e., the subventricular zone (SVZ) and the dentate gyrus of the hippocampus]. Insults to the brain such as cerebral ischemia result in a physiological mobilization of endogenous neural stem cells. Since recent studies showed that pharmacological stimulation can be used to expand the endogenous neural stem cell niche, hope has been raised to enhance the brain's own regenerative capacity. For the evaluation of such novel therapeutic approaches, longitudinal and intraindividual monitoring of the endogenous neural stem cell niche would be required. However, to date no conclusive imaging technique has been established. We used positron emission tomography (PET) and the radiotracer 3'-deoxy-3'-[(18)F]fluoro-l-thymidine ([(18)F]FLT) that enables imaging and measuring of proliferation to noninvasively detect endogenous neural stem cells in the normal and diseased adult rat brain in vivo. This method indeed visualized neural stem cell niches in the living rat brain, identified as increased [(18)F]FLT-binding in the SVZ and the hippocampus. Focal cerebral ischemia and subsequent damage of the blood-brain barrier did not interfere with the capability of [(18)F]FLT-PET to visualize neural stem cell mobilization. Moreover, [(18)F]FLT-PET allowed for an in vivo quantification of increased neural stem cell mobilization caused by pharmacological stimulation or by focal cerebral ischemia. The data suggest that noninvasive longitudinal monitoring and quantification of endogenous neural stem cell activation in the brain is feasible and that [(18)F]FLT-PET could be used to monitor the effects of drugs aimed at expanding the neural stem cell niche.


Asunto(s)
Neuronas/fisiología , Tomografía de Emisión de Positrones/métodos , Células Madre/fisiología , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Encéfalo/metabolismo , Encéfalo/fisiología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Didesoxinucleósidos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Insulina/farmacología , Péptidos y Proteínas de Señalización Intracelular , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/fisiología , Proteínas de la Membrana/farmacología , Neuronas/metabolismo , Ratas , Células Madre/metabolismo
14.
Cancer Res ; 67(4): 1706-15, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17308112

RESUMEN

To further develop gene therapy for patients with glioblastomas, an experimental gene therapy protocol was established comprising a series of imaging parameters for (i) noninvasive assessment of viable target tissue followed by (ii) targeted application of herpes simplex virus type 1 (HSV-1) amplicon vectors and (iii) quantification of treatment effects by imaging. We show that viable target tissue amenable for application of gene therapy vectors can be identified by multitracer positron emission tomography (PET) using 2-(18)F-fluoro-2-deoxy-D-glucose, methyl-(11)C-L-methionine, or 3'-deoxy-3'-(18)F-fluoro-L-thymidine ([(18)F]FLT). Targeted application of HSV-1 amplicon vectors containing two therapeutic genes with synergistic antitumor activity (Escherichia coli cytosine deaminase, cd, and mutated HSV-1 thymidine kinase, tk39, fused to green fluorescent protein gene, gfp) leads to an overall response rate of 68%, with 18% complete responses and 50% partial responses. Most importantly, we show that the "tissue dose" of HSV-1 amplicon vector-mediated gene expression can be noninvasively assessed by 9-[4-(18)F-fluoro-3-(hydroxymethyl)butyl]guanine ([(18)F]FHBG) PET. Therapeutic effects could be monitored by PET with significant differences in [(18)F]FLT accumulation in all positive control tumors and 72% in vivo transduced tumors (P = 0.01) as early as 4 days after prodrug therapy. For all stably and in vivo transduced tumors, cdIREStk39gfp gene expression as measured by [(18)F]FHBG-PET correlated with therapeutic efficiency as measured by [(18)F]FLT-PET. These data indicate that imaging-guided vector application with determination of tissue dose of vector-mediated gene expression and correlation to induced therapeutic effect using multimodal imaging is feasible. This strategy will help in the development of safe and efficient gene therapy protocols for clinical application.


Asunto(s)
Terapia Genética/métodos , Glioma/diagnóstico por imagen , Glioma/terapia , Animales , Línea Celular Tumoral , Didesoxinucleósidos , Radioisótopos de Flúor , Glioma/genética , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Ratones , Ratones Desnudos , Tomografía de Emisión de Positrones/métodos , Radiofármacos , Ratas , Ratas Desnudas
15.
J Tissue Eng Regen Med ; 13(6): 960-972, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30815982

RESUMEN

In the brain, neural stem cells (NSC) are tightly regulated by external signals and biophysical cues mediated by the local microenvironment or "niche." In particular, the influence of tissue elasticity, known to fundamentally affect the function of various cell types in the body, on NSC remains poorly understood. We, accordingly, aimed to characterize the effects of elastic substrates on critical NSC functions. Primary rat NSC were grown as monolayers on polydimethylsiloxane- (PDMS-) based gels. PDMS-coated cell culture plates, simulating the physiological microenvironment of the living brain, were generated in various degrees of elasticity, ranging from 1 to 50 kPa; additionally, results were compared with regular glass plates as usually used in cell culture work. Survival of NSC on the PDMS-based substrates was unimpaired. The proliferation rate on 1 kPa PDMS decreased by 45% compared with stiffer PMDS substrates of 50 kPa (p < 0.05) whereas expression of cyclin-dependent kinase inhibitor 1B/p27Kip1 increased more than two fold (p < 0.01), suggesting NSC quiescence. NSC differentiation was accelerated on softer substrates and favored the generation of neurons (42% neurons on 1 kPa PDMS vs. 25% on 50 kPa PDMS; p < 0.05). Neurons generated on 1 kPa PDMS showed 29% longer neurites compared with those on stiffer PDMS substrates (p < 0.05), suggesting optimized neuronal maturation and an accelerated generation of neuronal networks. Data show that primary NSC are significantly affected by the mechanical properties of their microenvironment. Culturing NSC on a substrate of brain-like elasticity keeps them in their physiological, quiescent state and increases their neurogenic potential.


Asunto(s)
Fenómenos Biofísicos , Encéfalo/fisiología , Elasticidad , Células-Madre Neurales/citología , Neurogénesis , Animales , Bovinos , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Proyección Neuronal , Ratas Wistar , Regulación hacia Arriba
16.
Front Cell Neurosci ; 13: 461, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31708742

RESUMEN

Despite its extensive use in clinical studies, the molecular mechanisms underlying the effects of transcranial direct current stimulation (tDCS) remain to be elucidated. We previously described subacute effects of tDCS on immune- and stem cells in the rat brain. To investigate the more immediate effects of tDCS regulating those cellular responses, we treated rats with a single session of either anodal or cathodal tDCS, and analyzed the gene expression by microarray; sham-stimulated rats served as control. Anodal tDCS increased expression of several genes coding for the major histocompatibility complex I (MHC I), while cathodal tDCS increased the expression of the immunoregulatory protein osteopontin (OPN). We confirmed the effects of gene upregulation by immunohistochemistry at the protein level. Thus, our data show a novel mechanism for the actions of tDCS on immune- and inflammatory processes, providing a target for future therapeutic studies.

17.
Neurosci Lett ; 434(3): 253-9, 2008 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-18337003

RESUMEN

Facial nerve lesions are common in humans and often require surgical intervention. If repair is delayed, reinnervation can be facilitated by transposing the freshly cut hypoglossal nerve end-to-end directly to the distal facial nerve, allowing for uncompromised hypoglossal axons to reinnervate the denervated facial musculature (hypoglossal-facial anastomosis, HFA). Schwann cells (SCs) in the distal nerve stump have an important function in promoting axonal regeneration by expressing multiple regeneration-associated proteins. Chronically denervated SCs cease to express those factors, but it is unknown whether they can be reactivated by fresh axonal sprouts and regain part of their function. We evaluated SC function and viability in distal facial nerve stump of rats at various time points after chronic denervation as well as following immediate or delayed HFA by assessing their expression of growth-associated protein 43 kDa (GAP-43) and the neuregulin receptors erbB2 and erbB4. Our results show that maximal upregulation of those factors in denervated SCs occurred a few weeks after nerve transection, indicating that a short period of denervation might even be beneficial before nerve repair. Motor SCs denervated for 32 weeks had downregulated their activity and ceased to express the regeneration-associated factors. SCs immediately re-expressed GAP-43, erbB2, and erbB4 following contact with fresh hypoglossal motor axons, demonstrating they are competent to promote regeneration even after long-term denervation.


Asunto(s)
Traumatismos del Nervio Facial/cirugía , Nervio Facial/fisiología , Neuronas Motoras/fisiología , Regeneración Nerviosa/fisiología , Transferencia de Nervios/métodos , Células de Schwann/fisiología , Animales , Atrofia/fisiopatología , Atrofia/prevención & control , Atrofia/cirugía , Biomarcadores/metabolismo , Desnervación , Receptores ErbB/metabolismo , Nervio Facial/citología , Nervio Facial/cirugía , Traumatismos del Nervio Facial/fisiopatología , Femenino , Proteína GAP-43/metabolismo , Glicoproteínas/metabolismo , Conos de Crecimiento/fisiología , Conos de Crecimiento/ultraestructura , Nervio Hipogloso/citología , Nervio Hipogloso/fisiología , Nervio Hipogloso/cirugía , Inmunohistoquímica , Neuronas Motoras/citología , Neurregulina-1/metabolismo , Ratas , Ratas Wistar , Receptor ErbB-2 , Receptor ErbB-4 , Recuperación de la Función/fisiología , Células de Schwann/citología , Factores de Tiempo
18.
Clin Cancer Res ; 13(22 Pt 1): 6761-8, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18006778

RESUMEN

PURPOSE: Lentiviral vectors pseudotyped with glycoproteins of the lymphocytic choriomeningitis virus (LCMV-GP) are promising candidates for gene therapy of malignant glioma, as they specifically and efficiently transduce glioma cells in vitro and in vivo. Here, we evaluated the therapeutic efficacy of LCMV-GP and vesicular stomatitis virus glycoprotein (VSV-G) pseudotyped vectors. EXPERIMENTAL DESIGN: Therapeutic efficacy was tested for unmodified (9L) and DsRed-modified (9LDsRed) gliomas using the suicide gene thymidine kinase of the herpes simplex virus type 1 (HSV-1-tk). Positron emission tomography (PET) and magnetic resonance imaging were done to analyze transduction of tumors and monitor therapeutic outcome. RESULTS: LCMV-GP pseudotypes mediated a successful eradication of 9LDsRed tumors with 100% of long-term survivors. Before initiation of ganciclovir treatment, a strong HSV-1-tk expression within the tumor was detected by noninvasive PET using the tracer 9-[4-[(18)F]fluoro-3-(hydroxymethyl)butyl]guanine. Therapeutic outcome was successfully monitored by magnetic resonance imaging and PET imaging and correlated with the histopathologic data. In the 9L model, LCMV-GP and VSV-G pseudotyped lentiviral vectors displayed similar therapeutic efficacy. Further studies revealed that normal brain cells transduced with VSV-G pseudotypes were not eliminated by ganciclovir treatment and contributed significantly to the bystander killing of tumor cells. CONCLUSIONS: Suicide gene transfer using pseudotyped lentiviral vectors was very effective in the treatment of rat glioma and therefore is an attractive therapeutic strategy also in human glioblastoma especially in conjunction with an imaging-guided approach. In addition, high selectivity of gene transfer to tumor cells may not always be desirable for therapeutic genes that exert a clear bystander effect.


Asunto(s)
Neoplasias Encefálicas/terapia , Genes Transgénicos Suicidas , Terapia Genética , Vectores Genéticos/genética , Glioma/terapia , Lentivirus/genética , Animales , Antivirales/farmacología , Encéfalo/citología , Ganciclovir/farmacología , Glicoproteínas/genética , Herpesvirus Humano 1/genética , Humanos , Virus de la Coriomeningitis Linfocítica/genética , Imagen por Resonancia Magnética , Tomografía de Emisión de Positrones , Ratas , Timidina Quinasa/genética , Vesiculovirus/genética
19.
Stem Cell Res Ther ; 9(1): 182, 2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29973246

RESUMEN

BACKGROUND: Osteopontin (OPN), an acidic phosphoglycoprotein, is upregulated in the brain after cerebral ischemia. We previously reported that OPN supports migration, survival, and proliferation of neural stem cells (NSC) in primary cell culture, as well as their differentiation into neurons. We here analyzed the effects of OPN on neuroblasts in vivo in the context of cerebral ischemia. METHODS: Transgenic mice expressing luciferase under the control of the neuroblast-specific doublecortin (DCX)-promoter, allowing visualization of neuroblasts in vivo using bioluminescence imaging (BLI), were injected with OPN intracerebroventricularly while control mice were injected with vehicle buffer. To assess the effects of OPN after ischemia, additional mice were subjected to photothrombosis and injected with either OPN or vehicle. RESULTS: OPN enhanced the migration of neuroblasts both in the healthy brain and after ischemia, as quantified by BLI in vivo. Moreover, the number of neural progenitors was increased following OPN treatment, with the maximum effect on the second day after OPN injection into the healthy brain, and 14 days after OPN injection following ischemia. After ischemia, OPN quantitatively promoted the endogenous, ischemia-induced neuroblast expansion, and additionally recruited progenitors from the contralateral hemisphere. CONCLUSIONS: Our results strongly suggest that OPN constitutes a promising substance for the targeted activation of neurogenesis in ischemic stroke.


Asunto(s)
Encéfalo/diagnóstico por imagen , Neurogénesis/efectos de los fármacos , Osteopontina/farmacología , Accidente Cerebrovascular/diagnóstico por imagen , Animales , Diferenciación Celular/fisiología , Movimiento Celular/efectos de los fármacos , Proteína Doblecortina , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Transgénicos
20.
Lab Anim ; 50(3): 212-6, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26442519

RESUMEN

Transcranial direct current stimulation (tDCS) constitutes a promising approach for promoting recovery of function after stroke, although the underlying neurobiological mechanisms are unclear. To conduct translational research in animal models, stimulation parameters should not lead to neuronal lesions. Liebetanz et al. recommend charge densities for cathodal stimulation in rats, but parameters for mice are not established. We established tDCS in the wild-type mouse, enabling studies with genetically-engineered mice (GEM). tDCS equipment was adapted to fit the mouse skull. Using different polarities and charge densities, tDCS was safe to apply in the mouse where the charge density was below 198 kC/m(2) for single or repeated stimulations. These findings are crucial for future investigations of the neurobiological mechanisms underlying tDCS using GEM.


Asunto(s)
Encéfalo/patología , Recuperación de la Función , Accidente Cerebrovascular/terapia , Estimulación Transcraneal de Corriente Directa/métodos , Animales , Modelos Animales de Enfermedad , Electrodos , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA