Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(11)2023 May 23.
Article in English | MEDLINE | ID: mdl-37298071

ABSTRACT

The apolipoprotein E (ApoE) gene is a genetic risk factor for late-onset Alzheimer's disease, in which ε4 allele carriers have increased risk compared to the common ε3 carriers. Cadmium (Cd) is a toxic heavy metal and a potential neurotoxicant. We previously reported a gene-environment interaction (GxE) effect between ApoE4 and Cd that accelerates or increases the severity of the cognitive decline in ApoE4-knockin (ApoE4-KI) mice exposed to 0.6 mg/L CdCl2 through drinking water compared to control ApoE3-KI mice. However, the mechanisms underlying this GxE effect are not yet defined. Because Cd impairs adult neurogenesis, we investigated whether genetic and conditional stimulation of adult neurogenesis can functionally rescue Cd-induced cognitive impairment in ApoE4-KI mice. We crossed either ApoE4-KI or ApoE3-KI to an inducible Cre mouse strain, Nestin-CreERTM:caMEK5-eGFPloxP/loxP (designated as caMEK5), to generate ApoE4-KI:caMEK5 and ApoE3-KI:caMEK5. Tamoxifen administration in these mice genetically and conditionally induces the expression of caMEK5 in adult neural stem/progenitor cells, enabling the stimulation of adult neurogenesis in the brain. Male ApoE4-KI:caMEK5 and ApoE3-KI:caMEK5 mice were exposed to 0.6 mg/L CdCl2 throughout the experiment, and tamoxifen was administered once Cd-induced impairment in spatial working memory was consistently observed. Cd exposure impaired spatial working memory earlier in ApoE4-KI:caMEK5 than in ApoE3-KI:caMEK5 mice. In both strains, these deficits were rescued after tamoxifen treatment. Consistent with these behavioral findings, tamoxifen treatment enhanced adult neurogenesis by increasing the morphological complexity of adult-born immature neurons. These results provide evidence for a direct link between impaired spatial memory and adult neurogenesis in this GxE model.


Subject(s)
Alzheimer Disease , Apolipoprotein E4 , Mice , Animals , Male , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Cadmium/metabolism , Mice, Transgenic , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Neurogenesis , Memory Disorders/metabolism , Hippocampus/metabolism , Tamoxifen/pharmacology , Tamoxifen/metabolism , Apolipoproteins E/metabolism , Alzheimer Disease/metabolism
2.
Environ Toxicol ; 37(2): 335-348, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34741586

ABSTRACT

Cadmium (Cd) is a toxic heavy metal and a significant public health concern. Epidemiological studies suggest that Cd is a potential neurotoxicant, and its exposure is associated with cognitive deficits in children, adults, and seniors. Our previous study has found that adulthood-only Cd exposure can impair cognition in mice. However, few studies have addressed the effects of Cd exposure during adolescence on cognitive behavior in animals later in life. In the present study, we exposed 4-week-old male C57BL/6 mice to 3 mg/L Cd via drinking water for 28 weeks and assessed their hippocampus-dependent learning and memory. Cd did not affect anxiety or locomotor activity in the open field test. However, Cd exposure impaired short-term spatial memory and contextual fear memory in mice. A separate cohort of 4-week-old mice was similarly exposed to Cd for 13 weeks to investigate the potential mechanism of Cd neurotoxicity on cognition. We observed that Cd-treated mice had fewer adult-born cells, adult-born neurons, and a reduced proportion of adult-born cells that differentiated into mature neurons in the subgranular zone of the dentate gyrus. These results suggest that Cd exposure from adolescence to adulthood is sufficient to cause cognitive deficits and impair key processes of hippocampal neurogenesis in mice.


Subject(s)
Cadmium , Memory , Animals , Cadmium/toxicity , Cognition , Hippocampus , Male , Mice , Mice, Inbred C57BL , Neurogenesis
3.
Neurotoxicology ; 81: 127-136, 2020 12.
Article in English | MEDLINE | ID: mdl-33039505

ABSTRACT

Cadmium (Cd) is a heavy metal that is one of the most toxic environmental pollutants throughout the world. We previously reported that Cd exposure impairs olfactory memory in mice. However, the underlying mechanisms for its neurotoxicity for olfactory function are not well understood. Since adult Subventricular zone (SVZ) and Olfactory Bulb (OB) neurogenesis contributes to olfaction, olfactory memory defects caused by Cd may be due to inhibition of neurogenesis. In this study, using bromodeoxyuridine (BrdU) labeling and immunohistochemistry, we found that 0.6 mg/L Cd exposure through drinking water impaired adult SVZ/OB neurogenesis in C57BL/6 mice. To determine if the inhibition of olfactory memory by Cd can be reversed by stimulating adult neurogenesis, we utilized the transgenic caMEK5 mouse strain to conditional stimulate of adult neurogenesis by activating the endogenous ERK5 MAP kinase signaling pathway. This was accomplished by conditionally induced expression of active MEK5 (caMEK5) in adult neural stem/progenitor cells. The caMEK5 mice were exposed to 0.6 mg/L Cd for 38 weeks, and tamoxifen was administered to induce caMEK5 expression and stimulate adult SVZ/OB neurogenesis during Cd exposure. Short-term olfactory memory test and sand-digging based, odor-cued olfactory learning and memory test were conducted after Cd and tamoxifen treatments to examine their effects on olfaction. Here we report that Cd exposure impaired short-term olfactory memory and odor-cued associative learning and memory in mice. Furthermore, the Cd-impaired olfactory memory deficits were rescued by the tamoxifen-induction of caMEK5 expression. This suggests that Cd exposure impairs olfactory function by affecting adult SVZ/OB neurogenesis in mice.


Subject(s)
Behavior, Animal , Lateral Ventricles/enzymology , Memory , Mitogen-Activated Protein Kinase 7/metabolism , Neurogenesis , Olfaction Disorders/prevention & control , Olfactory Bulb/enzymology , Olfactory Perception , Smell , Animals , Association Learning , Cadmium Chloride , Cues , Disease Models, Animal , Enzyme Activation , Female , Lateral Ventricles/pathology , Lateral Ventricles/physiopathology , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinase 7/genetics , Odorants , Olfaction Disorders/chemically induced , Olfaction Disorders/enzymology , Olfaction Disorders/physiopathology , Olfactory Bulb/pathology , Olfactory Bulb/physiopathology , Time Factors
4.
Toxicol Sci ; 177(1): 263-280, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32617577

ABSTRACT

Cadmium (Cd) is a heavy metal and an environmental pollutant. However, the full spectrum of its neurotoxicity and the underlying mechanisms are not completely understood. Our previous studies demonstrated that Cd exposure impairs adult hippocampal neurogenesis and hippocampus-dependent memory in mice. This study aims to determine if these adverse effects of Cd exposure can be mitigated by genetically and conditionally enhancing adult neurogenesis. To address this issue, we utilized the transgenic constitutive active MEK5 (caMEK5) mouse strain we previously developed and characterized. This mouse strain enables us to genetically and conditionally activate adult neurogenesis by administering tamoxifen to induce expression of a caMEK5 in adult neural stem/progenitor cells, which stimulates adult neurogenesis through activation of the endogenous extracellular signal-regulated kinase 5 mitogen-activated protein kinase pathway. The caMEK5 mice were exposed to 0.6 mg/l Cd through drinking water for 38 weeks. Once impairment of memory was confirmed, tamoxifen was administered to induce caMEK5 expression and to activate adult neurogenesis. Behavior tests were conducted at various time points to monitor hippocampus-dependent memory. Upon completion of the behavior tests, brain tissues were collected for cellular studies of adult hippocampal neurogenesis. We report here that Cd impaired hippocampus-dependent spatial memory and contextual fear memory in mice. These deficits were rescued by the tamoxifen induction of caMEK5 expression. Furthermore, Cd inhibition of adult hippocampal neurogenesis was also reversed. This rescue experiment provides strong evidence for a direct link between Cd-induced impairments of adult hippocampal neurogenesis and hippocampus-dependent memory.


Subject(s)
Cadmium , Neurogenesis , Animals , Cadmium/toxicity , Female , Hippocampus , Male , Memory , Mice , Mice, Inbred C57BL , Mice, Transgenic
5.
Toxicol Sci ; 173(1): 189-201, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31626305

ABSTRACT

Cadmium (Cd) is a heavy metal of great public health concern. Recent studies suggested a link between Cd exposure and cognitive decline in humans. The ε4 allele, compared with the common ε3 allele, of the human apolipoprotein E gene (ApoE) is associated with accelerated cognitive decline and increased risks for Alzheimer's disease (AD). To investigate the gene-environment interactions (GxE) between ApoE-ε4 and Cd exposure on cognition, we used a mouse model of AD that expresses human ApoE-ε3 (ApoE3-KI [knock-in]) or ApoE-ε4 (ApoE4-KI). Mice were exposed to 0.6 mg/l CdCl2 through drinking water for 14 weeks and assessed for hippocampus-dependent memory. A separate cohort was sacrificed immediately after exposure and used for Cd measurements and immunostaining. The peak blood Cd was 0.3-0.4 µg/l, within levels found in the U.S. general population. All Cd-treated animals exhibited spatial working memory deficits in the novel object location test. This deficit manifested earlier in ApoE4-KI mice than in ApoE3-KI within the same sex and earlier in males than females within the same genotype. ApoE4-KI but not ApoE3-KI mice exhibited reduced spontaneous alternation later in life in the T-maze test. Finally, Cd exposure impaired neuronal differentiation of adult-born neurons in the hippocampus of male ApoE4-KI mice. These data suggest that a GxE between ApoE4 and Cd exposure leads to accelerated cognitive impairment and that impaired adult hippocampal neurogenesis may be one of the underlying mechanisms. Furthermore, male mice were more susceptible than female mice to this GxE effect when animals were young.


Subject(s)
Apolipoprotein E4/metabolism , Cadmium/toxicity , Hazardous Substances/toxicity , Memory/drug effects , Alzheimer Disease , Animals , Apolipoprotein E3 , Behavior, Animal , Brain , Cognition , Disease Models, Animal , Female , Gene-Environment Interaction , Hippocampus , Humans , Male , Maze Learning , Memory Disorders , Mice , Mice, Transgenic
6.
Toxicol Sci ; 171(2): 501-514, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31271426

ABSTRACT

Cadmium (Cd) is an environmental pollutant of considerable interest throughout the world and potentially a neurotoxicant. Our recent data indicate that Cd exposure induces impairment of hippocampus-dependent learning and memory in mice. However, the underlying mechanisms for this defect are not known. The goal of this study was to determine if Cd inhibits adult neurogenesis and to identify underlying signaling pathways responsible for this impairment. Adult hippocampal neurogenesis is a process in which adult neural progenitor/stem cells (aNPCs) in the subgranular zone (SGZ) of the dentate gyrus (DG) generate functional new neurons in the hippocampus which contributes to hippocampus-dependent learning and memory. However, studies concerning the effects of neurotoxicants on adult hippocampal neurogenesis and the underlying signaling mechanisms are limited. Here, we report that Cd significantly induces apoptosis, inhibits proliferation, and impairs neuronal differentiation in primary cultured aNPCs derived from the SGZ. In addition, the c-Jun NH2-terminal kinase and p38 mitogen-activated protein kinase signaling pathways are activated by Cd and contribute to its toxicity. Furthermore, we exposed 8-week-old male C57BL/6 mice to Cd through drinking water for 13 weeks to assess the effects of Cd on adult hippocampal neurogenesis in vivo. Cd treatment reduced the number of 5-week-old adult-born cells in the DG and impaired the differentiation of adult-born hippocampal neurons. These results suggest that Cd exposure impairs adult hippocampal neurogenesis both in vitro and in vivo. This may contribute to Cd-mediated inhibition of hippocampus-dependent learning and memory.

7.
Toxicol Sci ; 161(1): 87-102, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29029324

ABSTRACT

Cadmium (Cd) is a heavy metal of high interest to the superfund initiative. Recent epidemiology studies have suggested a possible association between Cd exposure and cognitive as well as olfactory impairments in humans. However, studies in animal models are needed to establish a direct causal relationship between Cd exposure and impairments in cognition and olfaction. This study aims to investigate the toxic effect of Cd on cognition and olfactory function in mice. One group of 8-week-old C57BL/6 male mice was exposed to 3 mg/l Cd (in the form of CdCl2) through drinking water for 20 weeks for behavior tests and final blood Cd concentration analysis. The behavior tests were conducted before, during, and after Cd exposure to analyze the effects of Cd on cognition and olfactory function. Upon completion of behavior tests, blood was collected to measure final blood Cd concentration. Two additional groups of mice were similarly exposed to Cd for 5 or 13 weeks for peak blood Cd concentration measurement. The peak blood Cd concentration was 2.125-2.25 µg/l whereas the final blood Cd concentration was 0.18 µg/l. At this exposure level, Cd impaired hippocampus-dependent learning and memory in novel object location test, T-maze test, and contextual fear memory test. It also caused deficits in short-term olfactory memory and odor-cued olfactory learning and memory. Results in this study demonstrate a direct relationship between Cd exposure and cognitive as well as olfactory impairments in an animal model.


Subject(s)
Behavior, Animal/drug effects , Cadmium/toxicity , Cognitive Dysfunction/chemically induced , Environmental Pollutants/toxicity , Memory/drug effects , Olfaction Disorders/chemically induced , Animals , Cadmium/blood , Disease Models, Animal , Environmental Pollutants/blood , Hippocampus/drug effects , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL
8.
eNeuro ; 2(2)2015.
Article in English | MEDLINE | ID: mdl-26464972

ABSTRACT

Although there is evidence that adult neurogenesis contributes to the therapeutic efficacy of chronic antidepressant treatment for anxiety and depression disorders, the role of adult neurogenesis in the onset of depression-related symptoms is still open to question. To address this issue, we utilized a transgenic mouse strain in which adult neurogenesis was specifically and conditionally impaired by Nestin-CreER-driven, inducible knockout (icKO) of erk5 MAP kinase in Nestin-expressing neural progenitors of the adult mouse brain upon tamoxifen administration. Here, we report that inhibition of adult neurogenesis by this mechanism is not associated with an increase of the baseline anxiety or depression in non-stressed animals, nor does it increase the animal's susceptibility to depression after chronic unpredictable stress treatment. Our findings indicate that impaired adult neurogenesis does not lead to anxiety or depression.

9.
J Neurosci ; 35(20): 7833-49, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25995470

ABSTRACT

Recent discoveries have suggested that adult neurogenesis in the subventricular zone (SVZ) and olfactory bulb (OB) may be required for at least some forms of olfactory behavior in mice. However, it is unclear whether conditional and selective enhancement of adult neurogenesis by genetic approaches is sufficient to improve olfactory function under physiological conditions or after injury. Furthermore, specific signaling mechanisms regulating adult neurogenesis in the SVZ/OB are not fully defined. We previously reported that ERK5, a MAP kinase selectively expressed in the neurogenic regions of the adult brain, plays a critical role in adult neurogenesis in the SVZ/OB. Using a site-specific knock-in mouse model, we report here that inducible and targeted activation of the endogenous ERK5 in adult neural stem/progenitor cells enhances adult neurogenesis in the OB by increasing cell survival and neuronal differentiation. This conditional ERK5 activation also improves short-term olfactory memory and odor-cued associative olfactory learning under normal physiological conditions. Furthermore, these mice show enhanced recovery of olfactory function and have more adult-born neurons after a zinc sulfate-induced lesion of the main olfactory epithelium. We conclude that ERK5 MAP kinase is an important endogenous signaling pathway regulating adult neurogenesis in the SVZ/OB, and that conditional activation of endogenous ERK5 is sufficient to enhance adult neurogenesis in the OB thereby improving olfactory function both under normal conditions and after injury.


Subject(s)
Mitogen-Activated Protein Kinase 7/metabolism , Neurogenesis , Neurons/metabolism , Olfactory Bulb/metabolism , Smell , Animals , Cells, Cultured , Memory , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 7/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Neurons/cytology , Neurons/physiology , Olfactory Bulb/cytology , Olfactory Bulb/growth & development , Olfactory Bulb/physiology , Signal Transduction
10.
J Neurosci ; 34(6): 2130-47, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24501354

ABSTRACT

Recent studies have shown that inhibition of adult neurogenesis impairs the formation of hippocampus-dependent memory. However, it is not known whether increasing adult neurogenesis affects the persistence of hippocampus-dependent long-term memory. Furthermore, signaling mechanisms that regulate adult neurogenesis are not fully defined. We recently reported that the conditional and targeted knock-out of ERK5 MAP kinase in adult neurogenic regions of the mouse brain attenuates adult neurogenesis in the hippocampus and disrupts several forms of hippocampus-dependent memory. Here, we developed a gain-of-function knock-in mouse model to specifically activate endogenous ERK5 in the neurogenic regions of the adult brain. We report that the selective and targeted activation of ERK5 increases adult neurogenesis in the dentate gyrus by enhancing cell survival, neuronal differentiation, and dendritic complexity. Conditional ERK5 activation also improves the performance of challenging forms of spatial learning and memory and extends hippocampus-dependent long-term memory. We conclude that enhancing signal transduction of a single signaling pathway within adult neural stem/progenitor cells is sufficient to increase adult neurogenesis and improve the persistence of hippocampus-dependent memory. Furthermore, activation of ERK5 may provide a novel therapeutic target to improve long-term memory.


Subject(s)
Hippocampus/enzymology , Memory, Long-Term/physiology , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Neurogenesis/physiology , Age Factors , Animals , Animals, Newborn , Cell Differentiation/physiology , Enzyme Activation/physiology , Gene Knock-In Techniques , Hippocampus/cytology , Maze Learning/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic
11.
PLoS One ; 8(4): e61948, 2013.
Article in English | MEDLINE | ID: mdl-23630619

ABSTRACT

Recent studies have led to the exciting idea that adult-born neurons in the olfactory bulb (OB) may be critical for complex forms of olfactory behavior in mice. However, signaling mechanisms regulating adult OB neurogenesis are not well defined. We recently reported that extracellular signal-regulated kinase (ERK) 5, a MAP kinase, is specifically expressed in neurogenic regions within the adult brain. This pattern of expression suggests a role for ERK5 in the regulation of adult OB neurogenesis. Indeed, we previously reported that conditional deletion of erk5 in adult neurogenic regions impairs several forms of olfactory behavior in mice. Thus, it is important to understand how ERK5 regulates adult neurogenesis in the OB. Here we present evidence that shRNA suppression of ERK5 in adult neural stem/progenitor cells isolated from the subventricular zone (SVZ) reduces neurogenesis in culture. By contrast, ectopic activation of endogenous ERK5 signaling via expression of constitutive active MEK5, an upstream activating kinase for ERK5, stimulates neurogenesis. Furthermore, inducible and conditional deletion of erk5 specifically in the neurogenic regions of the adult mouse brain interferes with cell cycle exit of neuroblasts, impairs chain migration along the rostral migratory stream and radial migration into the OB. It also inhibits neuronal differentiation and survival. These data suggest that ERK5 regulates multiple aspects of adult OB neurogenesis and provide new insights concerning signaling mechanisms governing adult neurogenesis in the SVZ-OB axis.


Subject(s)
Cell Movement , Cell Survival , Mitogen-Activated Protein Kinase 7/genetics , Neurogenesis , Neurons/physiology , Olfactory Bulb/cytology , Animals , Cell Cycle , Cells, Cultured , Cerebral Ventricles/cytology , Cerebral Ventricles/enzymology , Gene Knockout Techniques , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 7/deficiency , Neural Stem Cells/physiology , Primary Cell Culture
12.
J Biol Chem ; 288(4): 2623-31, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23223235

ABSTRACT

Prolactin-stimulated adult neurogenesis in the subventricular zone (SVZ) and olfactory bulb (OB) mediates several reproductive behaviors including mating/pregnancy, dominant male pheromone preference in females, and paternal recognition of offspring. However, downstream signaling mechanisms underlying prolactin-induced adult neurogenesis are completely unknown. We report here for the first time that prolactin activates extracellular signal-regulated kinase 5 (ERK5), a MAP kinase that is specifically expressed in the neurogenic regions of the adult mouse brain. Knockdown of ERK5 by retroviral infection of shRNA attenuates prolactin-stimulated neurogenesis in SVZ-derived adult neural stem/progenitor cells (aNPCs). Inducible erk5 deletion in adult neural stem cells of transgenic mice inhibits neurogenesis in the SVZ and OB following prolactin infusion or mating/pregnancy. These results identify ERK5 as a novel and critical signaling mechanism underlying prolactin-induced adult neurogenesis.


Subject(s)
Brain/metabolism , Mitogen-Activated Protein Kinase 7/physiology , Olfactory Bulb/metabolism , Prolactin/metabolism , Animals , Brain Mapping/methods , Female , Gene Deletion , Genotype , Mice , Mice, Knockout , Microscopy, Confocal/methods , Mitogen-Activated Protein Kinase 7/metabolism , Neurogenesis , Recombinant Proteins/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Tamoxifen/pharmacology
13.
J Biol Chem ; 287(28): 23306-17, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22645146

ABSTRACT

Recent studies have led to the exciting idea that adult-born neurons in the dentate gyrus of the hippocampus may play a role in hippocampus-dependent memory formation. However, signaling mechanisms that regulate adult hippocampal neurogenesis are not well defined. Here we report that extracellular signal-regulated kinase 5 (ERK5), a member of the mitogen-activated protein kinase family, is selectively expressed in the neurogenic regions of the adult mouse brain. We present evidence that shRNA suppression of ERK5 in adult hippocampal neural stem/progenitor cells (aNPCs) reduces the number of neurons while increasing the number of cells expressing markers for stem/progenitor cells or proliferation. Furthermore, shERK5 attenuates both transcription and neuronal differentiation mediated by Neurogenin 2, a transcription factor expressed in adult hippocampal neural progenitor cells. By contrast, ectopic activation of endogenous ERK5 signaling via expression of constitutive active MEK5, an upstream activating kinase for ERK5, promotes neurogenesis in cultured aNPCs and in the dentate gyrus of the mouse brain. Moreover, neurotrophins including NT3 activate ERK5 and stimulate neuronal differentiation in aNPCs in an ERK5-dependent manner. Finally, inducible and conditional deletion of ERK5 specifically in the neurogenic regions of the adult mouse brain delays the normal progression of neuronal differentiation and attenuates adult neurogenesis in vivo. These data suggest ERK5 signaling as a critical regulator of adult hippocampal neurogenesis.


Subject(s)
Hippocampus/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Animals , Antineoplastic Agents, Hormonal/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Dentate Gyrus/metabolism , Hippocampus/cytology , Hippocampus/growth & development , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , MAP Kinase Kinase 5/genetics , MAP Kinase Kinase 5/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Mitogen-Activated Protein Kinase 7/genetics , NIH 3T3 Cells , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , RNA Interference , Tamoxifen/pharmacology
14.
J Neuropathol Exp Neurol ; 69(5): 511-20, 2010 May.
Article in English | MEDLINE | ID: mdl-20418776

ABSTRACT

Mechanistic studies underlying dopaminergic neuron death may identify new drug targets for the treatment of Parkinson disease. Epidemiological studies have linked pesticide exposure to increased risk for sporadic Parkinson disease. Here, we investigated the role of c-Jun-N-terminal kinase 3 (JNK3), a neural-specific JNK isoform, in dopaminergic neuron death induced by the pesticides rotenone and paraquat. The role of JNK3 was evaluated using RNA silencing and gene deletion to block JNK3 signaling. Using an antibody that recognizes all isoforms of activated JNKs, we found that paraquat and rotenone stimulate JNK phosphorylation in primary cultured dopaminergic neurons. In cultured neurons transfected with Jnk3-specific siRNA and in neurons from Jnk3 mice, JNK phosphorylation was nearly abolished, suggesting that JNK3 is the main JNK isoform activated in dopaminergic neurons by these pesticides. Paraquat- and rotenone-induced death of dopaminergic neurons was also significantly reduced by Jnk3 siRNA or Jnk3 gene deletion, and deletion of the Jnk3 gene completely attenuated paraquat-induced dopaminergic neuron death and motor deficits in vivo. Our data identify JNK3 as a common and critical mediator of dopaminergic neuron death induced by paraquat and rotenone, suggesting that it is a potential drug target for Parkinson disease treatment.


Subject(s)
Dopamine/metabolism , Herbicides/pharmacology , Insecticides/pharmacology , Mitogen-Activated Protein Kinase 10/metabolism , Neurons/drug effects , Paraquat/pharmacology , Rotenone/pharmacology , Animals , Caspase 3/metabolism , Cell Death/drug effects , Cell Death/genetics , Cells, Cultured , Dose-Response Relationship, Drug , Embryo, Mammalian , Exploratory Behavior/drug effects , Gene Deletion , Mesencephalon/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 10/deficiency , Mitogen-Activated Protein Kinase 10/genetics , Oxidative Stress/genetics , Phosphorylation/drug effects , RNA, Small Interfering/pharmacology , Reaction Time/drug effects , Time Factors , Tyrosine 3-Monooxygenase/metabolism
15.
PLoS One ; 4(4): e5204, 2009.
Article in English | MEDLINE | ID: mdl-19365559

ABSTRACT

The commitment of multi-potent cortical progenitors to a neuronal fate depends on the transient induction of the basic-helix-loop-helix (bHLH) family of transcription factors including Neurogenin 1 (Neurog1). Previous studies have focused on mechanisms that control the expression of these proteins while little is known about whether their pro-neural activities can be regulated by kinase signaling pathways. Using primary cultures and ex vivo slice cultures, here we report that both the transcriptional and pro-neural activities of Neurog1 are regulated by extracellular signal-regulated kinase (ERK) 5 signaling in cortical progenitors. Activation of ERK5 potentiated, while blocking ERK5 inhibited Neurog1-induced neurogenesis. Furthermore, endogenous ERK5 activity was required for Neurog1-initiated transcription. Interestingly, ERK5 activation was sufficient to induce Neurog1 phosphorylation and ERK5 directly phosphorylated Neurog1 in vitro. We identified S179/S208 as putative ERK5 phosphorylation sites in Neurog1. Mutations of S179/S208 to alanines inhibited the transcriptional and pro-neural activities of Neurog1. Our data identify ERK5 phosphorylation of Neurog1 as a novel mechanism regulating neuronal fate commitment of cortical progenitors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Mitogen-Activated Protein Kinase 7/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Enzyme Activation , Humans , Intermediate Filament Proteins/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 7/genetics , Nerve Tissue Proteins/genetics , Nestin , Neurons/cytology , Neurons/physiology , Phosphorylation , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stem Cells/cytology , Stem Cells/physiology , Transcription, Genetic
16.
Proc Natl Acad Sci U S A ; 103(25): 9697-702, 2006 Jun 20.
Article in English | MEDLINE | ID: mdl-16766652

ABSTRACT

Multipotent cortical progenitor cells differentiate into neurons and glial cells during development; however, mechanisms governing the specification of progenitors to a neuronal fate are not well understood. Although both extrinsic and intrinsic factors regulate this process, little is known about kinase signaling mechanisms that direct neuronal fate. Here, we report that extracellular signal-regulated kinase (ERK) 5 is expressed and active in proliferating cortical progenitors. Lentiviral gene delivery of a dominant negative ERK5 or dominant negative MAP kinase kinase 5 reduced the number of neurons generated from rat cortical progenitor cells in culture, whereas constitutive activation of ERK5 increased the production of neurons. Furthermore, when cortical progenitor cells were treated with ciliary neurotrophic factor, which induces precocious glial differentiation, ERK5 activation still promoted neuronal fate while suppressing glial differentiation. Our data also indicate that ERK5 does not directly regulate proliferation or apoptosis of cultured cortical progenitors. We conclude that ERK5 is necessary and sufficient to stimulate the generation of neurons from cortical progenitors. These results suggest a previously uncharacterized function for ERK5 signaling during brain development and raise the interesting possibility that extrinsic factors may instruct cortical progenitors to become neurons by activating the ERK5 pathway.


Subject(s)
Cerebellar Cortex/cytology , Cerebellar Cortex/enzymology , Mitogen-Activated Protein Kinase 7/metabolism , Neurons/cytology , Neurons/enzymology , Animals , Cell Adhesion , Cell Differentiation , Cell Proliferation , Cells, Cultured , Gene Expression Regulation, Enzymologic , Mitogen-Activated Protein Kinase 7/genetics , Neuroglia/cytology , Neuroglia/enzymology , Rats , Rats, Sprague-Dawley , Stem Cells/cytology , Stem Cells/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...