Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Nature ; 619(7970): 606-615, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37438521

ABSTRACT

The specific loss of midbrain dopamine neurons (mDANs) causes major motor dysfunction in Parkinson's disease, which makes cell replacement a promising therapeutic approach1-4. However, poor survival of grafted mDANs remains an obstacle to successful clinical outcomes5-8. Here we show that the surgical procedure itself (referred to here as 'needle trauma') triggers a profound host response that is characterized by acute neuroinflammation, robust infiltration of peripheral immune cells and brain cell death. When midbrain dopamine (mDA) cells derived from human induced pluripotent stem (iPS) cells were transplanted into the rodent striatum, less than 10% of implanted tyrosine hydroxylase (TH)+ mDANs survived at two weeks after transplantation. By contrast, TH- grafted cells mostly survived. Notably, transplantation of autologous regulatory T (Treg) cells greatly modified the response to needle trauma, suppressing acute neuroinflammation and immune cell infiltration. Furthermore, intra-striatal co-transplantation of Treg cells and human-iPS-cell-derived mDA cells significantly protected grafted mDANs from needle-trauma-associated death and improved therapeutic outcomes in rodent models of Parkinson's disease with 6-hydroxydopamine lesions. Co-transplantation with Treg cells also suppressed the undesirable proliferation of TH- grafted cells, resulting in more compact grafts with a higher proportion and higher absolute numbers of TH+ neurons. Together, these data emphasize the importance of the initial inflammatory response to surgical injury in the differential survival of cellular components of the graft, and suggest that co-transplanting autologous Treg cells effectively reduces the needle-trauma-induced death of mDANs, providing a potential strategy to achieve better clinical outcomes for cell therapy in Parkinson's disease.


Subject(s)
Cell- and Tissue-Based Therapy , Dopaminergic Neurons , Graft Survival , Neuroinflammatory Diseases , Parkinson Disease , T-Lymphocytes, Regulatory , Tyrosine 3-Monooxygenase , Humans , Dopamine/analogs & derivatives , Dopamine/metabolism , Dopaminergic Neurons/immunology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/transplantation , Mesencephalon/pathology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/prevention & control , Neuroinflammatory Diseases/therapy , Parkinson Disease/complications , Parkinson Disease/pathology , Parkinson Disease/surgery , Parkinson Disease/therapy , Tyrosine 3-Monooxygenase/deficiency , Tyrosine 3-Monooxygenase/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Cell- and Tissue-Based Therapy/methods , Animals , Mice , Rats , Oxidopamine/metabolism , Graft Survival/immunology , Cell Death , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Neostriatum/metabolism , Time Factors , Cell Proliferation , Treatment Outcome
2.
N Engl J Med ; 382(20): 1926-1932, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32402162

ABSTRACT

We report the implantation of patient-derived midbrain dopaminergic progenitor cells, differentiated in vitro from autologous induced pluripotent stem cells (iPSCs), in a patient with idiopathic Parkinson's disease. The patient-specific progenitor cells were produced under Good Manufacturing Practice conditions and characterized as having the phenotypic properties of substantia nigra pars compacta neurons; testing in a humanized mouse model (involving peripheral-blood mononuclear cells) indicated an absence of immunogenicity to these cells. The cells were implanted into the putamen (left hemisphere followed by right hemisphere, 6 months apart) of a patient with Parkinson's disease, without the need for immunosuppression. Positron-emission tomography with the use of fluorine-18-L-dihydroxyphenylalanine suggested graft survival. Clinical measures of symptoms of Parkinson's disease after surgery stabilized or improved at 18 to 24 months after implantation. (Funded by the National Institutes of Health and others.).


Subject(s)
Dopaminergic Neurons/cytology , Induced Pluripotent Stem Cells/transplantation , Parkinson Disease/therapy , Pars Compacta/cytology , Aged , Animals , Basal Ganglia/diagnostic imaging , Basal Ganglia/metabolism , Cell Differentiation , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/transplantation , Follow-Up Studies , Humans , Induced Pluripotent Stem Cells/immunology , Male , Mice , Mice, SCID , Parkinson Disease/diagnostic imaging , Positron-Emission Tomography , Putamen/diagnostic imaging , Tomography, X-Ray Computed , Transplantation, Autologous , Transplantation, Homologous
3.
Nat Chem Biol ; 16(8): 876-886, 2020 08.
Article in English | MEDLINE | ID: mdl-32451509

ABSTRACT

The orphan nuclear receptor Nurr1 is critical for the development, maintenance and protection of midbrain dopaminergic (mDA) neurons. Here we show that prostaglandin E1 (PGE1) and its dehydrated metabolite, PGA1, directly interact with the ligand-binding domain (LBD) of Nurr1 and stimulate its transcriptional function. We also report the crystallographic structure of Nurr1-LBD bound to PGA1 at 2.05 Å resolution. PGA1 couples covalently to Nurr1-LBD by forming a Michael adduct with Cys566, and induces notable conformational changes, including a 21° shift of the activation function-2 helix (H12) away from the protein core. Furthermore, PGE1/PGA1 exhibit neuroprotective effects in a Nurr1-dependent manner, prominently enhance expression of Nurr1 target genes in mDA neurons and improve motor deficits in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned mouse models of Parkinson's disease. Based on these results, we propose that PGE1/PGA1 represent native ligands of Nurr1 and can exert neuroprotective effects on mDA neurons, via activation of Nurr1's transcriptional function.


Subject(s)
Alprostadil/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Prostaglandins A/metabolism , Animals , Cell Line, Tumor , Crystallography, X-Ray , Dopamine/metabolism , Humans , Ligands , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurons/metabolism , Neuroprotective Agents/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 2/chemistry , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Protein Binding , Rats , Signal Transduction , Transcription, Genetic
4.
Brain ; 138(Pt 10): 3030-47, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26133660

ABSTRACT

Aberrant glutathione or Ca(2+) homeostasis due to oxidative stress is associated with the pathogenesis of neurodegenerative disorders. The Ca(2+)-permeable transient receptor potential cation (TRPC) channel is predominantly expressed in the brain, which is sensitive to oxidative stress. However, the role of the TRPC channel in neurodegeneration is not known. Here, we report a mechanism of TRPC5 activation by oxidants and the effect of glutathionylated TRPC5 on striatal neurons in Huntington's disease. Intracellular oxidized glutathione leads to TRPC5 activation via TRPC5 S-glutathionylation at Cys176/Cys178 residues. The oxidized glutathione-activated TRPC5-like current results in a sustained increase in cytosolic Ca(2+), activated calmodulin-dependent protein kinase and the calpain-caspase pathway, ultimately inducing striatal neuronal cell death. We observed an abnormal glutathione pool indicative of an oxidized state in the striatum of Huntington's disease transgenic (YAC128) mice. Increased levels of endogenous TRPC5 S-glutathionylation were observed in the striatum in both transgenic mice and patients with Huntington's disease. Both knockdown and inhibition of TRPC5 significantly attenuated oxidation-induced striatal neuronal cell death. Moreover, a TRPC5 blocker improved rearing behaviour in Huntington's disease transgenic mice and motor behavioural symptoms in littermate control mice by increasing striatal neuron survival. Notably, low levels of TRPC1 increased the formation of TRPC5 homotetramer, a highly Ca(2+)-permeable channel, and stimulated Ca(2+)-dependent apoptosis in Huntington's disease cells (STHdh(Q111/111)). Taken together, these novel findings indicate that increased TRPC5 S-glutathionylation by oxidative stress and decreased TRPC1 expression contribute to neuronal damage in the striatum and may underlie neurodegeneration in Huntington's disease.


Subject(s)
Corpus Striatum/pathology , Glutathione/metabolism , Green Fluorescent Proteins/metabolism , Huntington Disease/pathology , Neurons/metabolism , TRPC Cation Channels/metabolism , Analysis of Variance , Animals , Calcium/metabolism , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Huntingtin Protein , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/genetics , RNA, Small Interfering/metabolism , TRPC Cation Channels/genetics , Transfection
5.
Cell Res ; 34(7): 479-492, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38777859

ABSTRACT

Parkinson's disease (PD) stands as the second most common neurodegenerative disorder after Alzheimer's disease, and its prevalence continues to rise with the aging global population. Central to the pathophysiology of PD is the specific degeneration of midbrain dopamine neurons (mDANs) in the substantia nigra. Consequently, cell replacement therapy (CRT) has emerged as a promising treatment approach, initially supported by various open-label clinical studies employing fetal ventral mesencephalic (fVM) cells. Despite the initial favorable results, fVM cell therapy has intrinsic and logistical limitations that hinder its transition to a standard treatment for PD. Recent efforts in the field of cell therapy have shifted its focus towards the utilization of human pluripotent stem cells, including human embryonic stem cells and induced pluripotent stem cells, to surmount existing challenges. However, regardless of the transplantable cell sources (e.g., xenogeneic, allogeneic, or autologous), the poor and variable survival of implanted dopamine cells remains a major obstacle. Emerging evidence highlights the pivotal role of host immune responses following transplantation in influencing the survival of implanted mDANs, underscoring an important area for further research. In this comprehensive review, building upon insights derived from previous fVM transplantation studies, we delve into the functional ramifications of host immune responses on the survival and efficacy of grafted dopamine cells. Furthermore, we explore potential strategic approaches to modulate the host immune response, ultimately aiming for optimal outcomes in future clinical applications of CRT for PD.


Subject(s)
Cell- and Tissue-Based Therapy , Parkinson Disease , Humans , Parkinson Disease/therapy , Cell- and Tissue-Based Therapy/methods , Animals , Dopaminergic Neurons/metabolism , Stem Cell Transplantation
6.
Nat Protoc ; 17(3): 890-909, 2022 03.
Article in English | MEDLINE | ID: mdl-35140411

ABSTRACT

To fully realize the potential of human pluripotent stem cells (hPSCs) for both therapeutic and research purposes, it is critical to follow an efficient and reliable in vitro differentiation method that is based on optimal physical, chemical and developmental cues. This highly reproducible protocol describes how to grow hPSCs such as human induced pluripotent and embryonic stem cells in a physically confined area ('spot') and efficiently differentiate them into a highly enriched population of healthy and functional midbrain dopamine progenitors (mDAPs) and midbrain dopamine neurons (mDANs). The protocol takes 28 d, during which cells first grow and differentiate in spots for 14 d and then are replated and further differentiated for a further 14 d as a monolayer culture. We describe how to produce mDAPs, control the quality of cells and cryopreserve mDAPs without loss of viability. Previously we showed that mDANs generated by this 'spotting'-based method exhibit gene expression and (electro)physiological properties typical of A9 mDANs lost in Parkinson's disease brains and can rescue motor defects when transplanted into the striatum of 6-hydroxydopamine-lesioned rats. This protocol is scalable for production of mDAPs under good manufacturing practice conditions and was also previously successfully used to generate cells for the first autologous cell replacement therapy of a patient with Parkinson's disease without the need for immune suppression. We anticipate this protocol could also be readily adapted to use spotting-based culture to further optimize the differentiation of hPSC to alternative differentiated cell types.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Animals , Cell Differentiation , Dopamine/metabolism , Dopaminergic Neurons , Humans , Mesencephalon , Rats
7.
Cell Rep ; 37(13): 110155, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34965411

ABSTRACT

During somatic reprogramming, cellular energy metabolism fundamentally switches from predominantly mitochondrial oxidative phosphorylation toward glycolysis. This metabolic reprogramming, also called the Warburg effect, is critical for the induction of pluripotency, but its molecular mechanisms remain poorly defined. Notably, SIRT2 is consistently downregulated during the reprogramming process and regulates glycolytic switch. Here, we report that downregulation of SIRT2 increases acetylation of mitogen-activated protein kinase (MAPK) kinase-1 (MEK1) at Lys175, resulting in activation of extracellular signal-regulated kinases (ERKs) and subsequent activation of the pro-fission factor dynamin-related protein 1 (DRP1). In parallel, downregulation of SIRT2 hyperacetylates the serine/threonine protein kinase AKT1 at Lys20 in a non-canonical way, activating DRP1 and metabolic reprogramming. Together, our study identified two axes, SIRT2-MEK1-ERK-DRP1 and SIRT2-AKT1-DRP1, that critically link mitochondrial dynamics and oxidative phosphorylation to the somatic reprogramming process. These upstream signals, together with SIRT2's role in glycolytic switching, may underlie the Warburg effect observed in human somatic cell reprogramming.


Subject(s)
Cellular Reprogramming , Dynamins/metabolism , MAP Kinase Kinase 1/metabolism , Mitochondrial Dynamics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sirtuin 2/metabolism , Acetylation , Dynamins/genetics , Energy Metabolism , Fibroblasts/metabolism , Glycolysis , Humans , MAP Kinase Kinase 1/genetics , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Oxidative Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Sirtuin 2/genetics
8.
Biomol Ther (Seoul) ; 29(5): 483-491, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34045367

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder that involves the loss of dopaminergic neurons in the substantia nigra (SN). Matrix metalloproteinases-8 (MMP-8), neutrophil collagenase, is a functional player in the progressive pathology of various inflammatory disorders. In this study, we administered an MMP-8 inhibitor (MMP-8i) in Leucine-rich repeat kinase 2 (LRRK2) G2019S transgenic mice, to determine the effects of MMP-8i on PD pathology. We observed a significant increase of ionized calcium- binding adapter molecule 1 (Iba1)-positive activated microglia in the striatum of LRRK2 G2019S mice compared to normal control mice, indicating enhanced neuro-inflammatory responses. The increased number of Iba1-positive activated microglia in LRRK2 G2019S PD mice was down-regulated by systemic administration of MMP-8i. Interestingly, this LRRK2 G2019S PD mice showed significantly reduced size of cell body area of tyrosine hydroxylase (TH) positive neurons in SN region and MMP-8i significantly recovered cellular atrophy shown in PD model indicating distinct neuro-protective effects of MMP-8i. Furthermore, MMP-8i administration markedly improved behavioral abnormalities of motor balancing coordination in rota-rod test in LRRK2 G2019S mice. These data suggest that MMP-8i attenuates the pathological symptoms of PD through anti-inflammatory processes.

9.
J Clin Invest ; 130(2): 904-920, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31714896

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder associated with loss of striatal dopamine, secondary to degeneration of midbrain dopamine (mDA) neurons in the substantia nigra, rendering cell transplantation a promising therapeutic strategy. To establish human induced pluripotent stem cell-based (hiPSC-based) autologous cell therapy, we report a platform of core techniques for the production of mDA progenitors as a safe and effective therapeutic product. First, by combining metabolism-regulating microRNAs with reprogramming factors, we developed a method to more efficiently generate clinical-grade iPSCs, as evidenced by genomic integrity and unbiased pluripotent potential. Second, we established a "spotting"-based in vitro differentiation methodology to generate functional and healthy mDA cells in a scalable manner. Third, we developed a chemical method that safely eliminates undifferentiated cells from the final product. Dopaminergic cells thus express high levels of characteristic mDA markers, produce and secrete dopamine, and exhibit electrophysiological features typical of mDA cells. Transplantation of these cells into rodent models of PD robustly restores motor function and reinnervates host brain, while showing no evidence of tumor formation or redistribution of the implanted cells. We propose that this platform is suitable for the successful implementation of human personalized autologous cell therapy for PD.


Subject(s)
Induced Pluripotent Stem Cells , Motor Activity , Parkinson Disease, Secondary , Recovery of Function , Stem Cell Transplantation , Animals , Autografts , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Induced Pluripotent Stem Cells/transplantation , Male , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Parkinson Disease, Secondary/therapy , Rats , Rats, Nude
10.
Mol Neurobiol ; 55(2): 1607-1619, 2018 02.
Article in English | MEDLINE | ID: mdl-28190238

ABSTRACT

Alpha-synuclein (α-SYN) is expressed during neuronal development and is mainly involved in the modulation of synaptic transmission. Missense mutations and amplifications of this gene have been associated with the pathogenesis of Parkinson's disease. Here, we evaluate whether α-SYN plays a detrimental role in the phenotypic and morphological regulation of neurons. We also identify the underlying mechanisms of this process in all-trans-retinoic acid (RA)-induced differentiated SH-SY5Y cells, which represents dopaminergic (DAergic) phenotype. Our results indicate that overexpression of wild-type or mutant A53T α-SYN attenuated the RA-induced upregulation of tyrosine hydroxylase and dopamine transporter as well as neurite outgrowth in SH-SY5Y cells. In addition, GSK-3ß inactivation and downstream ß-catenin stabilization were associated with RA-induced differentiation, which was attenuated by α-SYN. Moreover, protein phosphatase 2A was positively regulated by α-SYN and was implicated in the α-SYN-mediated interference with RA signaling. The results obtained from SH-SY5Y cells were verified in primary cultures of mesencephalic DAergic neurons from A53T α-SYN transgenic mice, which represent high levels of α-SYN and protein phosphatase 2A in the midbrain. The number and length of neurites in tyrosine hydroxylase-positive as well as Tau-positive cells from A53T α-SYN transgenic mice were significantly lower than those in littermate controls. The current results provide novel insight into the role of α-SYN in the regulation of neuronal differentiation, including DAergic neurons. Identifying the signaling pathway involved in the α-SYN-mediated dysregulation of neuronal differentiation could lead to a better understanding of the developmental processes underlying α-SYN-related pathologies and facilitate the discovery of specifically targeted therapeutics.


Subject(s)
Cell Differentiation/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Neurons/drug effects , Signal Transduction/drug effects , Tretinoin/pharmacology , alpha-Synuclein/metabolism , beta Catenin/metabolism , Cell Line, Tumor , Dopamine Plasma Membrane Transport Proteins/metabolism , Humans , Neurites/drug effects , Neurites/metabolism , Neurons/metabolism , Tyrosine 3-Monooxygenase/metabolism , Up-Regulation/drug effects
11.
Aging (Albany NY) ; 9(3): 803-822, 2017 03 12.
Article in English | MEDLINE | ID: mdl-28288000

ABSTRACT

Expansion of polyglutamine stretch in the huntingtin (HTT) protein is a major cause of Huntington's disease (HD). The polyglutamine part in HTT interacts with various proteins implicated in epigenetic regulation of genes, suggesting that mutant HTT may disturb the integrity of the epigenetic system. Here, we used a PCRseq-based method to examine expression profile of 395 exonic segments from 260 "epi-driver" genes in splenic T lymphocytes from aged HD mice. We identified 67 exonic segments differentially expressed between young and aged HD mice, most of them upregulated in the aged. Polycomb-repressive complex (PRC)-regulated genes (PRGs) were markedly upregulated in aged HD mice, consistent with downregulation of PRC genes. Epi-driver gene categories of lysine-methylation, lysine-demethylation, arginine-methylation, and PRG showed differential age-associated changes between HD and control. Analyzing the pattern of change in epi-driver gene expressions hinted at an enhanced shift in HD chromatin to a more accessible state with age, which was experimentally demonstrated by DNase-I-hypersensitivity sequencing showing increased chromatin accessibility in HD cells compared to control. We suggest the global change can potentially relieve chromatin-induced repression of many genes, and the unintended expressions of some detrimental proteins could alter T cell function to a greater degree in aged HD mice.


Subject(s)
Chromatin/metabolism , Huntington Disease/genetics , Polycomb-Group Proteins/genetics , T-Lymphocytes/metabolism , Animals , Disease Models, Animal , Gene Expression Regulation , Huntington Disease/metabolism , Mice , Polycomb-Group Proteins/metabolism
12.
Mol Neurobiol ; 53(4): 2276-86, 2016 May.
Article in English | MEDLINE | ID: mdl-25976370

ABSTRACT

Huntington's disease (HD) is a movement disorder characterized by the early selective degeneration of striatum. For motor control, the striatum receives excitatory inputs from multiple brain regions and projects the information to other basal ganglia nuclei. Despite the pathological importance of the striatal degeneration in HD, there are little anatomical data that show impaired striatal connectivity in HD. For the anatomical mapping of the striatum, we injected here a neurotracer DiD to the dorsal striatum of HD mouse model (YAC128). Compared with littermate controls, the number of the traced inputs to the striatum was reduced dramatically in YAC128 mice at 12 months of age suggesting massive destruction of the striatal connections. Basal ganglia inputs were significantly damaged in HD mice by showing 61 % decrease in substantia nigra pars compacta, 85% decrease in thalamic centromedian nucleus, and 55% decrease in thalamic parafascicular nucleus. Cortical inputs were also greatly decreased by 43% in motor cortex, 48% in somatosensory cortex, and 72% in visual cortex. Besides the known striatal connections, the neurotracer DiD also traced inputs from amygdala and the amygdala inputs were decreased by 68% in YAC128 mice. Considering the role of amygdala in emotion processing, the impairment in amygdalostriatal connectivity strongly suggests that emotional disturbances could occur in HD mice. Indeed, open-field tests further indicated that YAC128 mice exhibited changes in emotional behaviors related to symptoms of depression and anxiety. Although onset of HD is clinically determined on the basis of motor abnormality, emotional deficits are also common features of the disease. Therefore, our anatomical connectivity mapping of the striatum provides a new insight to interpret brain dysfunction in HD.


Subject(s)
Corpus Striatum/pathology , Huntington Disease/pathology , Nerve Net/pathology , Animals , Behavior, Animal , Disease Models, Animal , Emotions , Mice , Neurons/pathology , Staining and Labeling
13.
Mol Neurobiol ; 53(1): 95-108, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25407931

ABSTRACT

Dysfunction of growth factor (GF) activities contributes to the decline and death of neurons during aging and in neurodegenerative diseases. In addition, neurons become more resistant to GF signaling with age. Micro (mi)RNAs are posttranscriptional regulators of gene expression that may be crucial to age- and disease-related changes in GF functions. MiR-126 is involved in regulating insulin/IGF-1/phosphatidylinositol-3-kinase (PI3K)/AKT and extracellular signal-regulated kinase (ERK) signaling, and we recently demonstrated a functional role of miR-126 in dopamine neuronal cell survival in models of Parkinson's disease (PD)-associated toxicity. Here, we show that elevated levels of miR-126 increase neuronal vulnerability to ubiquitous toxicity mediated by staurosporine (STS) or Alzheimer's disease (AD)-associated amyloid beta 1-42 peptides (Aß1-42). The neuroprotective factors IGF-1, nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and soluble amyloid precursor protein α (sAPPα) could diminish but not abrogate the toxic effects of miR-126. In miR-126 overexpressing neurons derived from Tg6799 familial AD model mice, we observed an increase in Aß1-42 toxicity, but surprisingly, both Aß1-42 and miR-126 promoted neurite sprouting. Pathway analysis revealed that miR-126 overexpression downregulated elements in the GF/PI3K/AKT and ERK signaling cascades, including AKT, GSK-3ß, ERK, their phosphorylation, and the miR-126 targets IRS-1 and PIK3R2. Finally, inhibition of miR-126 was neuroprotective against both STS and Aß1-42 toxicity. Our data provide evidence for a novel mechanism of regulating GF/PI3K signaling in neurons by miR-126 and suggest that miR-126 may be an important mechanistic link between metabolic dysfunction and neurotoxicity in general, during aging, and in the pathogenesis of specific neurological disorders, including PD and AD.


Subject(s)
Alzheimer Disease/metabolism , MicroRNAs/metabolism , Neurons/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cell Survival/physiology , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycogen Synthase Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rats, Sprague-Dawley
14.
Neurochem Int ; 69: 35-40, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24607701

ABSTRACT

Lipopolysaccharide (LPS) administration may be used to induce an in vivo model for neuroinflammation or neurodegeneration. We examined the regional distribution of inflammatory markers induced by LPS in the brain of young mice. Criteria for inflammation included measures of cytokines and microglial activation. Levels of IL-1ß mRNA increased in the frontal cortex, parietal cortex, hippocampus, and striatum following systemic treatment with LPS. Levels of SRA mRNA increased in the frontal cortex and striatum and levels of TLR2 and TLR4 mRNAs increased in the frontal cortex and cerebellum. Iba1-positive microglial cells increased in the striatum, medial septum, frontal cortex, and hippocampus after LPS treatment. In addition, glutathione (GSH) levels decreased and mitochondrial complex II/III activities increased after systemic LPS injection. Although LPS treatment did not significantly alter cellular ATP levels, these levels correlated with levels of IL-1ß and TLR4 in the LPS-treated mice. The region-specific inflammatory response to LPS in the brain may serve to create a model for studies of neurodegenerative disease.


Subject(s)
Brain/drug effects , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Microglia/drug effects , Mitochondria/drug effects , Animals , Brain/metabolism , Disease Models, Animal , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Male , Mice, Inbred ICR , Microglia/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Neurons/drug effects , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL