Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
Glia ; 71(10): 2383-2400, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37334743

RESUMO

The precise timing of neural progenitor development and the correct balance between proliferation and differentiation are crucial to generating a functional brain. The number, survival, and differentiation of neural progenitors during postnatal neurogenesis and gliogenesis is a highly regulated process. Postnatally, the majority of brain oligodendrocytes are generated from progenitors residing in the subventricular zone (SVZ), the germinal niche surrounding the lateral ventricles. In this study, we demonstrate that the p75 neurotrophin receptor (p75NTR) is highly expressed by OPCs in the postnatal male and female rat SVZ. Whereas the p75NTR is known to initiate apoptotic signaling after brain injury, it is highly expressed by proliferating progenitors in the SVZ, suggesting that it may have a different function during development. Lack of p75NTR reduced progenitor proliferation and caused premature oligodendrocyte differentiation and maturation both in vitro and in vivo, leading to aberrant early myelin formation. Our data reveal a novel role for p75NTR as a rheostat for oligodendrocyte production and maturation during myelin formation in the postnatal rat brain.


Assuntos
Ventrículos Laterais , Células Precursoras de Oligodendrócitos , Animais , Feminino , Masculino , Ratos , Encéfalo , Diferenciação Celular , Neurogênese/fisiologia , Oligodendroglia/fisiologia , Receptor de Fator de Crescimento Neural
2.
Dev Neurosci ; 2023 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-37348472

RESUMO

Hypoxic-ischemic (HI) brain injury in neonatal encephalopathy triggers a wave of neuroinflammatory events attributed to causing the progressive degeneration and functional deficits seen weeks after the primary damage. The cellular processes mediating this prolonged neurodegeneration in HI injury are not sufficiently understood. Consequently, current therapies are not fully protective. In a recent study, we found significant improvements in neurologic outcomes when a small molecule antagonist for Activin-Like Kinase 5 (ALK5), a transforming growth factor beta (TGF-ß) receptor was used as a therapeutic in a rat model of moderate term HI. Here, we have extended those studies to a mouse preterm pup model of HI. For these studies, postnatal day 7 (P7) CD1 mice of both sexes were exposed to 35-40 minutes of HI. Beginning 3 days later, SB505124, the ALK5 receptor antagonist, was administered systemically through intraperitoneal injections performed every 12 hours for 5 days. When evaluated 23 days later, SB505124-treated mice had ~2.5-fold more hippocampal area and ~2-fold more thalamic tissue. Approximately ~90% of the ipsilateral hemisphere (ILH) was preserved in the SB505124-treated HI mice compared to the vehicle-treated HI mice, where the ILH was ~60% of its normal size. SB505124 also preserved the subcortical white matter. SB505124 treatment preserved levels of aquaporin-4 and n-cadherin, key proteins associated with blood-brain-barrier function. Importantly, SB505124 administration improved sensorimotor function as assessed by a battery of behavioral tests. Altogether these data lend additional support to the conclusion that SB505124 is a candidate neuroprotective molecule that could be an effective treatment for HI-related encephalopathy in moderately injured preterm infants.

3.
J Neurosci Res ; 100(2): 578-597, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34811802

RESUMO

Traumatic brain injury (TBI) is a significant problem that affects over 800,000 children each year. As cell proliferation is disturbed by injury and required for normal brain development, we investigated how a pediatric closed head injury (CHI) would affect the progenitors of the subventricular zone (SVZ). Additionally, we evaluated the contribution of leukemia inhibitory factor (LIF) using germline LIF heterozygous mice (LIF Het), as LIF is an injury-induced cytokine, known to influence neurogenesis and gliogenesis. CHIs were performed on P20 LIF Het and wild-type (WT) mice. Ki-67 immunostaining and stereology revealed that cell proliferation increased ~250% in injured LIF Het mice compared to the 30% increase observed in injured WT mice at 48-hr post-CHI. OLIG2+ cell proliferation increased in the SVZ and white matter of LIF Het injured mice at 48-hr recovery. Using an 8-color flow cytometry panel, the proliferation of three distinct multipotential progenitors and early oligodendrocyte progenitor cell proliferation was significantly increased in LIF Het injured mice compared to WT injured mice. Supporting its cytostatic function, LIF decreased neurosphere progenitor and oligodendrocyte progenitor cell proliferation compared to controls. In highly enriched mouse oligodendrocyte progenitor cell cultures, LIF increased phospho-protein kinase B after 20 min and increased phospho-S6 ribosomal protein at 20 and 40 min of exposure, which are downstream targets of the mammalian target of rapamycin pathway. Altogether, our data provide new insights into the regulatory role of LIF in suppressing neural progenitor cell proliferation and, in particular, oligodendrocyte progenitor cell proliferation after a mild TBI.


Assuntos
Lesões Encefálicas Traumáticas , Células Precursoras de Oligodendrócitos , Animais , Proliferação de Células/fisiologia , Humanos , Fator Inibidor de Leucemia , Mamíferos , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/fisiologia
4.
Brain Behav Immun ; 101: 23-36, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34954074

RESUMO

Epidemiologic studies have demonstrated that infections during pregnancy increase the risk of offspring developing Schizophrenia, Autism, Depression and Bipolar Disorder and have implicated interleukin-6 (IL-6) as a causal agent. However, other cytokines have been associated with the developmental origins of psychiatric disorders; therefore, it remains to be established whether elevating IL-6 is sufficient to alter the trajectory of neural development. Furthermore, most rodent studies have manipulated the maternal immune system at mid-gestation, which affects the stem cells and progenitors in both the primary and secondary germinal matrices. Therefore, a question that remains to be addressed is whether elevating IL-6 when the secondary germinal matrices are most active will affect brain development. Here, we have increased IL-6 from postnatal days 3-6 when the secondary germinal matrices are rapidly expanding. Using Nestin-CreERT2 fate mapping we show that this transient increase in IL-6 decreased neurogenesis in the dentate gyrus of the dorsal hippocampus, reduced astrogliogenesis in the amygdala and decreased oligodendrogenesis in the body and splenium of the corpus callosum all by âˆ¼ 50%. Moreover, the IL-6 treatment elicited behavioral changes classically associated with neurodevelopmental disorders. As adults, IL-6 injected male mice lost social preference in the social approach test, spent âˆ¼ 30% less time socially engaging with sexually receptive females and produced âˆ¼ 50% fewer ultrasonic vocalizations during mating. They also engaged âˆ¼ 50% more time in self-grooming behavior and had an increase in inhibitory avoidance. Altogether, these data provide new insights into the biological mechanisms linking perinatal immune activation to complex neurodevelopmental brain disorders.


Assuntos
Interleucina-6 , Neurogênese , Comportamento Social , Animais , Comportamento Animal , Corpo Caloso , Citocinas , Feminino , Hipocampo/fisiologia , Masculino , Camundongos , Neurogênese/fisiologia , Gravidez
5.
Methods ; 133: 54-64, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-28887084

RESUMO

Currently, there is no widely accepted technique to efficiently and reproducibly grow stem and progenitor cells in vitro. Stem cells require contact with extracellular matrices as well as signals from growth factors to proliferate and to retain their stemness. We have shown a novel tissue culture platform (StemTrix cultureware) that transforms standard tissue culture plasticware into a multi-functional chitosan-based scaffold that supports the expansion of neural stem cells. The StemTrix scaffold is comprised of chitosan with immobilized heparin which in turn tethers heparin-binding growth factors. The scaffold is also coated with an adhesive ECM protein. Here we demonstrate that fibronectin or the RGD peptide contained in fibronectin are equally effective in promoting the adhesion, viability and growth of rat and human neural stem cells. When FGF-2 and heparin-binding EGF are tethered to the StemTrix cultureware neural stem cells grow ∼3 times faster and remain in a more primitive state as determined by both Western Blot and gene expression analyses. Another important feature of this new culture platform is that the NSCs remain in a primitive and proliferative state for 4days without refreshing the culture medium or providing new growth factors, which represents a 20-fold extension of FGF-2's biological activity vs when it is freely soluble in the medium. To test the utility of this scaffold for propagating other types of stem cells and progenitors we tethered platelet-derived growth factor (PDGF) and FGF-2 alone and in combination to the scaffold and tested the efficacy of this platform to maintain primary oligodendrocyte progenitors or the CG-4 cell line in a primitive state. Oligodendrocyte progenitors plated onto this multifunctional film proliferated for at least 3days without providing soluble growth factors while inhibiting the expression of the differentiation marker myelin-basic protein. Oligodendrocyte progenitors proliferated 3 times more rapidly than cells maintained on fibronectin-coated culture substrates in culture medium supplemented with soluble FGF-2 and PDGF. Finally, we show that StemTrix cultureware can be produced using clinical grade components, providing users with a fully defined platform suitable for clinical use that maintains stem cells or progenitors in a more uniform and primitive state.


Assuntos
Materiais Biocompatíveis/química , Células-Tronco Neurais/citologia , Transplante de Células-Tronco , Técnicas de Cultura de Tecidos/métodos , Animais , Materiais Biocompatíveis/farmacologia , Proteínas de Transporte/química , Proteínas de Transporte/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Quitosana/química , Quitosana/farmacologia , Citocinas/química , Citocinas/farmacologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fibronectinas/química , Fibronectinas/metabolismo , Humanos , Células-Tronco Neurais/efeitos dos fármacos , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Ratos , Alicerces Teciduais/química
6.
Ann Neurol ; 81(4): 560-571, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28253550

RESUMO

OBJECTIVE: Neonatal white matter injury (NWMI) is a lesion found in preterm infants that can lead to cerebral palsy. Although antagonists of bone morphogenetic protein (BMP) signaling, such as Noggin, promote oligodendrocyte precursor cell (OPC) production after hypoxic-ischemic (HI) injury, the downstream functional targets are poorly understood. The basic helix-loop-helix protein, oligodendrocyte transcription factor 1 (Olig1), promotes oligodendrocyte (OL) development and is essential during remyelination in adult mice. Here, we investigated whether Olig1 function is required downstream of BMP antagonism for response to injury in the neonatal brain. METHODS: We used wild-type and Olig1-null mice subjected to neonatal stroke and postnatal neural progenitor cultures, and we analyzed Olig1 expression in human postmortem samples from neonates that suffered HI encephalopathy (HIE). RESULTS: Olig1-null neonatal mice showed significant hypomyelination after moderate neonatal stroke. Surprisingly, damaged white matter tracts in Olig1-null mice lacked Olig2+ OPCs, and instead proliferating neuronal precursors and GABAergic interneurons were present. We demonstrate that Noggin-induced OPC production requires Olig1 function. In postnatal neural progenitors, Noggin governs production of OLs versus interneurons through Olig1-mediated repression of Dlx1/2 transcription factors. Additionally, we observed that Olig1 and the BMP signaling effector, phosphorylated SMADs (Sma- and Mad-related proteins) 1, 5, and 8, were elevated in the subventricular zone of human infants with HIE compared to controls. INTERPRETATION: These findings indicate that Olig1 has a critical function in regulation of postnatal neural progenitor cell production in response to Noggin. Ann Neurol 2017;81:560-571.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Oligodendroglia/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Técnicas de Cultura de Células , Modelos Animais de Doenças , Humanos , Lactente , Recém-Nascido , Doenças do Recém-Nascido , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neurais
7.
Pediatr Res ; 83(1-2): 385-396, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29028220

RESUMO

There is great interest in the regenerative potential of the neural stem cells and progenitors that populate the germinal zones of the immature brain. Studies using animal models of pediatric brain injuries have provided a clearer understanding of the responses of these progenitors to injury. In this review, we have compared and contrasted the responses of the endogenous neural stem cells and progenitors of the subventricular zone in animal models of neonatal cerebral hypoxia-ischemia, neonatal stroke, congenital cardiac disease, and pediatric traumatic brain injury. We have reviewed the dynamic shifts that occur within this germinal zone with injury as well as changes in known signaling molecules that affect these progenitors. Importantly, we have summarized data on the extent to which cell replacement occurs in response to each of these injuries, opportunities available, and obstacles that will need to be overcome to improve neurological outcomes in survivors.


Assuntos
Encefalopatias/terapia , Lesões Encefálicas Traumáticas/terapia , Lesões Encefálicas/terapia , Hipóxia-Isquemia Encefálica/fisiopatologia , Ventrículos Laterais/citologia , Células-Tronco Neurais/citologia , Animais , Animais Recém-Nascidos , Encéfalo/fisiopatologia , Diferenciação Celular , Ventrículos Cerebrais/fisiopatologia , Cardiopatias Congênitas/terapia , Humanos , Recém-Nascido , Camundongos , Neurônios/citologia , Oligodendroglia , Acidente Vascular Cerebral/terapia
9.
Dev Neurosci ; 39(1-4): 338-351, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28628913

RESUMO

Neonatal encephalopathy due to hypoxic-ischemic (HI) brain injury triggers a wave of neuroinflammatory events attributed to causing the progressive degeneration and functional deficits seen weeks after the initial insult. In a recent set of studies, we evaluated the therapeutic efficacy of a small molecule antagonist for ALK5 (activin-like kinase 5 ), TGF-ß receptor in a rat model of moderate perinatal HI and found significant improvements in neurologic outcomes. Here, we have extended those studies to evaluate the efficacy of delayed TGF-ß receptor antagonism on postnatal day (P) 6 and P9 HI rat pups with and without hypothermia. The ALK5 receptor antagonist SB505124 was administered systemically by osmotic pump beginning 3 days following HI. Extending our earlier data set that showed protection of the hippocampus in P6 pups treated with SB505124, these animals sustained less damage to their hippocampi and had improved performance on the Morris water maze (MWM) when tested on P60 versus vehicle-treated HI animals. By contrast, SB505124 did not improve sensorimotor deficits and exacerbated hippocampal and thalamic volume loss when administered 3 days after HI to P9 pups. SB505124-treated rats injured on P9 tended to perform worse than their vehicle-treated counterparts on MWM, and SB505124 treatment did not preserve hippocampal or thalamic neurons in P9 pups when combined with hypothermia. To elucidate the mechanism whereby ALK5 inhibition reduced neuronal death in the P6 HI model, we assessed levels of autophagy markers in neurons of the neocortex, hippocampus, and thalamus, and in the subcortical white matter, and found that SB505124 increased numbers of autophagosomes and levels of lipidated LC3 (light chain 3), a key protein known to mediate autophagy. Altogether, our results demonstrate that there is a dynamic switch in the CNS response to TGF-ß1 that occurs around P9 in rats where TGF-ß signaling inhibition worsens functional outcomes. This response is similar to the outcome of antagonizing TGF-ß signaling in adult stroke and other CNS disease models. We conclude that attenuating TGF-ß1 signaling will likely be an effective treatment for HI-related encephalopathy in moderately preterm infants, offering protection of the neocortex, hippocampus, and thalamus with enhanced cerebral autophagy contributing to the decrease in the extent of progressive neuronal cell death.


Assuntos
Benzodioxóis/farmacologia , Hipóxia-Isquemia Encefálica , Imidazóis/farmacologia , Fármacos Neuroprotetores/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Piridinas/farmacologia , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Fatores Etários , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Hipotermia Induzida , Distribuição Aleatória , Ratos , Ratos Wistar , Receptor do Fator de Crescimento Transformador beta Tipo I
10.
J Neurosci ; 35(23): 8855-65, 2015 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-26063918

RESUMO

Neonatal hypoxia-ischemia (H-I) is the leading cause of brain damage resulting from birth complications. Studies in neonatal rats have shown that H-I acutely expands the numbers of neural precursors (NPs) within the subventricular zone (SVZ). The aim of these studies was to establish which NPs expand after H-I and to determine how leukemia inhibitory factor (LIF) insufficiency affects their response. During recovery from H-I, the number of Ki67(+) cells in the medial SVZ of the injured hemisphere increased. Similarly, the number and size of primary neurospheres produced from the injured SVZ increased approximately twofold versus controls, and, upon differentiation, more than twice as many neurospheres from the damaged brain were tripotential, suggesting an increase in neural stem cells (NSCs). However, multimarker flow cytometry for CD133/LeX/NG2/CD140a combined with EdU incorporation revealed that NSC frequency diminished after H-I, whereas that of two multipotential progenitors and three unique glial-restricted precursors expanded, attributable to changes in their proliferation. By quantitative PCR, interleukin-6, LIF, and CNTF mRNA increased but with significantly different time courses, with LIF expression correlating best with NP expansion. Therefore, we evaluated the NP response to H-I in LIF-haplodeficient mice. Flow cytometry revealed that one subset of multipotential and bipotential intermediate progenitors did not increase after H-I, whereas another subset was amplified. Altogether, our studies demonstrate that neonatal H-I alters the composition of the SVZ and that LIF is a key regulator for a subset of intermediate progenitors that expand during acute recovery from neonatal H-I.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Regulação da Expressão Gênica/fisiologia , Hipóxia-Isquemia Encefálica/patologia , Células-Tronco Neurais/fisiologia , Neurônios/fisiologia , Animais , Animais Recém-Nascidos , Antígenos/metabolismo , Antígenos CD/metabolismo , Fator Neurotrófico Ciliar/genética , Fator Neurotrófico Ciliar/metabolismo , Modelos Animais de Doenças , Lateralidade Funcional , Hipóxia-Isquemia Encefálica/fisiopatologia , Antígeno Ki-67/metabolismo , Ventrículos Laterais/patologia , Fator Inibidor de Leucemia/metabolismo , Antígenos CD15/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/fisiologia , Proteoglicanas/metabolismo
11.
J Neurosci Res ; 94(12): 1531-1545, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27661001

RESUMO

Brain injuries, such as cerebral hypoxia-ischemia (H-I), induce a regenerative response from the neural stem/progenitors (NSPs) of the subventricular zone (SVZ); however, the mechanisms that regulate this expansion have not yet been fully elucidated. The Notch- Delta-Serrate-Lag2 (DSL) signaling pathway is considered essential for the maintenance of neural stem cells, but it is not known if it is necessary for the expansion of the NSPs subsequent to perinatal H-I injury. Therefore, the aim of this study was to investigate whether this pathway contributes to NSP expansion in the SVZ after H-I and, if so, to establish whether this pathway is directly induced by H-I or regulated by paracrine factors. Here we report that Notch1 receptor induction and one of its ligands, Delta-like 1, precedes NSP expansion after perinatal H-I in P6 rat pups and that this increase occurs specifically in the most medial cell layers of the SVZ where the stem cells reside. Pharmacologically inhibiting Notch signaling in vivo diminished NSP expansion. With an in vitro model of H-I, Notch1 was not induced directly by hypoxia, but was stimulated by soluble factors, specifically leukemia inhibitory factor, produced by astrocytes within the SVZ. These data confirm the importance both of the Notch-DSL signaling pathway in the expansion of NSPs after H-I and in the role of the support cells in their niche. They further support the body of evidence that indicates that leukemia inhibitory factor is a key injury-induced cytokine that is stimulating the regenerative response of the NSPs. © 2016 Wiley Periodicals, Inc.


Assuntos
Astrócitos/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Fator Inibidor de Leucemia/biossíntese , Regeneração Nervosa , Células-Tronco Neurais , Animais , Citocinas/metabolismo , Diaminas/farmacologia , Feminino , Ventrículos Laterais/patologia , Gravidez , Ratos , Ratos Wistar , Receptor Notch1/biossíntese , Receptor Notch1/genética , Receptores Opioides delta/biossíntese , Transdução de Sinais , Tiazóis/farmacologia
12.
J Biol Chem ; 289(8): 4626-33, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24398690

RESUMO

The objective of this study was to employ genetically engineered IGF-II analogs to establish which receptor(s) mediate the stemness promoting actions of IGF-II on mouse subventricular zone neural precursors. Neural precursors from the subventricular zone were propagated in vitro in culture medium supplemented with IGF-II analogs. Cell growth and identity were analyzed using sphere generation and further analyzed by flow cytometry. F19A, an analog of IGF-II that does not bind the IGF-2R, stimulated an increase in the proportion of neural stem cells (NSCs) while decreasing the proportion of the later stage progenitors at a lower concentration than IGF-II. V43M, which binds to the IGF-2R with high affinity but which has low binding affinity to the IGF-1R and to the A isoform of the insulin receptor (IR-A) failed to promote NSC growth. The positive effects of F19A on NSC growth were unaltered by the addition of a functional blocking antibody to the IGF-1R. Altogether, these data lead to the conclusion that IGF-II promotes stemness of NSCs via the IR-A and not through activation of either the IGF-1R or the IGF-2R.


Assuntos
Fator de Crescimento Insulin-Like II/análogos & derivados , Fator de Crescimento Insulin-Like II/farmacologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Receptor de Insulina/metabolismo , Animais , Biomarcadores/metabolismo , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Humanos , Proteína 2 Inibidora de Diferenciação/metabolismo , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica/efeitos dos fármacos , Receptor IGF Tipo 2/metabolismo , Esferoides Celulares/citologia , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo
13.
J Biol Chem ; 289(13): 9430-9, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24509861

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a member of the TNF family. The interaction of TRAIL with death receptor 4 (DR4) and DR5 can trigger apoptotic cell death. The aim of this study was to investigate the role of TRAIL signaling in neonatal hypoxia-ischemia (HI). Using a neonatal mouse model of HI, mRNA, and protein expression of TRAIL, DR5 and the TRAIL decoy receptors osteoprotegerin (OPG), mDcTRAILR1, and mDcTRAILR2 were determined. In vitro, mRNA expression of these genes was measured in primary neurons and oligodendrocyte progenitor cells (OPCs) after inflammatory cytokine (TNF-α/IFN-γ) treatment and/or oxygen and glucose deprivation (OGD). The toxicity of these various paradigms was also measured. The expression of TRAIL, DR5, OPG, and mDcTRAILR2 was significantly increased after HI. In vitro, inflammatory cytokines and OGD treatment significantly induced mRNAs for TRAIL, DR5, OPG, and mDcTRAILR2 in primary neurons and of TRAIL and OPG in OPCs. TRAIL protein was expressed primarily in microglia and astroglia, whereas DR5 co-localized with neurons and OPCs in vivo. OGD enhanced TNF-α/IFN-γ toxicity in both neuronal and OPC cultures. Recombinant TRAIL exerted toxicity alone or in combination with OGD and TNF-α/IFN-γ in primary neurons but not in OPC cultures. The marked increases in the expression of TRAIL and its receptors after cytokine exposure and OGD in primary neurons and OPCs were similar to those found in our animal model of neonatal HI. The toxicity of TRAIL in primary neurons suggests that TRAIL signaling participates in neonatal brain injury after inflammation and HI.


Assuntos
Sistema Nervoso Central/patologia , Hipóxia/patologia , Isquemia/patologia , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/deficiência , Hipóxia/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Interferon gama/farmacologia , Isquemia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/patologia , Oligodendroglia/patologia , Oxigênio/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Fator de Necrose Tumoral alfa/farmacologia
14.
Dev Neurosci ; 37(1): 29-42, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25377490

RESUMO

Pediatric traumatic brain injury is a significant problem that affects many children each year. Progress is being made in developing neuroprotective strategies to combat these injuries. However, investigators are a long way from therapies to fully preserve injured neurons and glia. To restore neurological function, regenerative strategies will be required. Given the importance of stem cells in repairing damaged tissues and the known persistence of neural precursors in the subventricular zone (SVZ), we evaluated regenerative responses of the SVZ to a focal brain lesion. As tissues repair more slowly with aging, injury responses of male Sprague Dawley rats at 6, 11, 17, and 60 days of age and C57Bl/6 mice at 14 days of age were compared. In the injured immature animals, cell proliferation in the dorsolateral SVZ more than doubled by 48 h. By contrast, the proliferative response was almost undetectable in the adult brain. Three approaches were used to assess the relative numbers of bona fide neural stem cells, as follows: the neurosphere assay (on rats injured at postnatal day 11, P11), flow cytometry using a novel 4-marker panel (on mice injured at P14) and staining for stem/progenitor cell markers in the niche (on rats injured at P17). Precursors from the injured immature SVZ formed almost twice as many spheres as precursors from uninjured age-matched brains. Furthermore, spheres formed from the injured brain were larger, indicating that the neural precursors that formed these spheres divided more rapidly. Flow cytometry revealed a 2-fold increase in the percentage of stem cells, a 4-fold increase in multipotential progenitor-3 cells and a 2.5-fold increase in glial-restricted progenitor-2/multipotential-3 cells. Analogously, there was a 2-fold increase in the mitotic index of nestin+/Mash1- immunoreactive cells within the immediately subependymal region. As the early postnatal SVZ is predominantly generating glial cells, an expansion of precursors might not necessarily lead to the production of many new neurons. On the contrary, many BrdU+/doublecortin+ cells were observed streaming out of the SVZ into the neocortex 2 weeks after injuries to P11 rats. However, very few new mature neurons were seen adjacent to the lesion 28 days after injury. Altogether, these data indicate that immature SVZ cells mount a more robust proliferative response to a focal brain injury than adult cells, which includes an expansion of stem cells, primitive progenitors and neuroblasts. Nonetheless, this regenerative response does not result in significant neuronal replacement, indicating that new strategies need to be implemented to retain the regenerated neurons and glia that are being produced.


Assuntos
Lesões Encefálicas/patologia , Proliferação de Células/fisiologia , Ventrículos Laterais/citologia , Células-Tronco Neurais/citologia , Neuroglia/citologia , Neurônios/citologia , Animais , Animais Recém-Nascidos , Contagem de Células/métodos , Diferenciação Celular , Ventrículos Cerebrais/fisiologia , Proteína Duplacortina , Masculino , Camundongos Endogâmicos C57BL , Ratos Sprague-Dawley
15.
J Neurochem ; 128(3): 376-90, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24032666

RESUMO

Isolating a pure population of neural stem cells (NSCs) has been difficult since no exclusive surface markers have been identified for panning or FACS purification. Moreover, additional refinements for maintaining NSCs in culture are required, since NSCs generate a variety of neural precursors (NPs) as they proliferate. Here, we demonstrate that post-natal rat NPs express low levels of pro-apoptotic molecules and resist phosphatidylinositol 3'OH kinase and extracellular regulated kinase 1/2 inhibition as compared to late oligodendrocyte progenitors. Furthermore, maintaining subventricular zone precursors in LY294002 and PD98059, inhibitors of PI3K and ERK1/2 signaling, eliminated lineage-restricted precursors as revealed by enrichment for Nestin(+)/SOX-2(+) cells. The cells that survived formed neurospheres and 89% of these neurospheres were tripotential, generating neurons, astrocytes, and oligodendrocytes. Without this enrichment step, less than 50% of the NPs were Nestin(+)/SOX-2(+) and 42% of the neurospheres were tripotential. In addition, neurospheres enriched using this procedure produced 3-times more secondary neurospheres, supporting the conclusion that this procedure enriches for NSCs. A number of genes that enhance survival were more highly expressed in neurospheres compared to late oligodendrocyte progenitors. Altogether, these studies demonstrate that primitive neural precursors can be enriched using a relatively simple and inexpensive means that will facilitate cell replacement strategies using stem cells as well as other studies whose goal is to reveal the fundamental properties of primitive neural precursors.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Animais , Western Blotting , Morte Celular/efeitos dos fármacos , Células Cultivadas , Ceramidas/toxicidade , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Análise em Microsséries , Nestina/metabolismo , Neuroglia/efeitos dos fármacos , Ensaios de Proteção de Nucleases , Oligodendroglia/efeitos dos fármacos , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína bcl-X/metabolismo
16.
Dev Neurosci ; 40(5-6): 381, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-31256168
17.
Stem Cells ; 30(6): 1265-76, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22593020

RESUMO

Insulin-like growth factor (IGF)-I and IGF-II regulate brain development and growth through the IGF type 1 receptor (IGF-1R). Less appreciated is that IGF-II, but not IGF-I, activates a splice variant of the insulin receptor (IR) known as IR-A. We hypothesized that IGF-II exerts distinct effects from IGF-I on neural stem/progenitor cells (NSPs) via its interaction with IR-A. Immunofluorescence revealed high IGF-II in the medial region of the subventricular zone (SVZ) comprising the neural stem cell niche, with IGF-II mRNA predominant in the adjacent choroid plexus. The IGF-1R and the IR isoforms were differentially expressed with IR-A predominant in the medial SVZ, whereas the IGF-1R was more abundant laterally. Similarly, IR-A was more highly expressed by NSPs, whereas the IGF-1R was more highly expressed by lineage restricted cells. In vitro, IGF-II was more potent in promoting NSP expansion than either IGF-I or standard growth medium. Limiting dilution and differentiation assays revealed that IGF-II was superior to IGF-I in promoting stemness. In vivo, NSPs propagated in IGF-II migrated to and took up residence in periventricular niches while IGF-I-treated NSPs predominantly colonized white matter. Knockdown of IR or IGF-1R using shRNAs supported the conclusion that the IGF-1R promotes progenitor proliferation, whereas the IR is important for self-renewal. Q-PCR revealed that IGF-II increased Oct4, Sox1, and FABP7 mRNA levels in NSPs. Our data support the conclusion that IGF-II promotes the self-renewal of neural stem/progenitors via the IR. By contrast, IGF-1R functions as a mitogenic receptor to increase precursor abundance.


Assuntos
Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Animais , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like II/genética , Camundongos , Transdução de Sinais
18.
Neurosci Lett ; 795: 137033, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36574812

RESUMO

The combination of lipopolysaccharide (LPS) and hypoxia-ischemia (HI) has been used to model the brain injury sustained by sick pre-term infants in order to study the pathological conditions of diffuse white matter injury, which is a major cause of preterm morbidity. Prior studies have shown that the timing and dose of LPS administration will determine whether the injury is reduced or exacerbated. Here we show that administering a single injection of LPS (0.1 mg/kg) to postnatal-day-2 rat pups 14 h before inducing HI effectively protects the brain from HI-associated damage. We show that the LPS-treated HI rat pups have significantly less histopathology compared to the saline-treated HI rat pups. Apoptotic deaths were dramatically curtailed in both the neocortex and white matter when evaluated at 2 days of recovery. Microglial activation was reduced when the percentage of CD68+/Iba1+ cells was quantified in the neocortex of the LPS-treated vs the saline-treated HI rat pups. One mechanism through which LPS pre-treatment appears to be preventing injury is through the AKT-endothelial nitric oxide synthase (eNOS) pathway as LPS induced an increase in both the expression and phosphorylation of eNOS. Altogether these data show that the neocortex, as well as the white matter sustain damage after HI at this timepoint in forebrain development and that acutely activating the immune system can protect the brain from brain injury.


Assuntos
Lesões Encefálicas , Hipóxia-Isquemia Encefálica , Fármacos Neuroprotetores , Animais , Ratos , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo , Animais Recém-Nascidos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Fármacos Neuroprotetores/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Inflamação/metabolismo , Encéfalo/metabolismo , Hipóxia/metabolismo , Isquemia/metabolismo , Lesões Encefálicas/metabolismo , Modelos Animais de Doenças
19.
Neurotrauma Rep ; 4(1): 236-250, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37095853

RESUMO

Leukemia inhibitory factor (LIF) is a neuroprotective cytokine that is essential for appropriate glial responses, remyelination, and preservation of neuronal conductance after injury. The intranasal route for delivery of therapeutics to the central nervous system is of particular interest given that it bypasses the blood-brain barrier and peripheral clearance systems. We explored the possibility that LIF might improve neurological function when administered intranasally during the acute phase in a pediatric model of mild traumatic brain injury (mTBI). We tested two doses of LIF and evaluated behavioral outcomes. Here, we show that acute 40-ng intranasal LIF treatment twice a day for 3 days attenuates astrogliosis and microgliosis, protects against axonal damage, significantly improves sensorimotor function, and is well tolerated without detrimental effects on growth. Altogether, our studies provide pre-clinical evidence for the use of acute intranasal LIF treatment as a viable therapeutic for pediatric cases of mTBIs.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa