Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
1.
Mol Autism ; 15(1): 10, 2024 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-38383466

RESUMO

BACKGROUND: A growing body of evidence suggests that immune dysfunction and inflammation in the peripheral tissues as well as the central nervous system are associated with the neurodevelopmental deficits observed in autism spectrum disorder (ASD). Elevated expression of pro-inflammatory cytokines in the plasma, serum, and peripheral blood mononuclear cells of ASD has been reported. These cytokine expression levels are associated with the severity of behavioral impairments and symptoms in ASD. In a prior study, our group reported that tumor necrosis factor-α (TNF-α) expression in granulocyte-macrophage colony-stimulating factor-induced macrophages (GM-CSF MΦ) and the TNF-α expression ratio in GM-CSF MΦ/M-CSF MΦ (macrophage colony-stimulating factor-induced macrophages) was markedly higher in individuals with ASD than in typically developed (TD) individuals. However, the mechanisms of how the macrophages and the highly expressed cytokines affect neurons remain to be addressed. METHODS: To elucidate the effect of macrophages on human neurons, we used a co-culture system of control human-induced pluripotent stem cell-derived neurons and differentiated macrophages obtained from the peripheral blood mononuclear cells of five TD individuals and five individuals with ASD. All participants were male and ethnically Japanese. RESULTS: Our results of co-culture experiments showed that GM-CSF MΦ affect the dendritic outgrowth of neurons through the secretion of pro-inflammatory cytokines, interleukin-1α and TNF-α. Macrophages derived from individuals with ASD exerted more severe effects than those derived from TD individuals. LIMITATIONS: The main limitations of our study were the small sample size with a gender bias toward males, the use of artificially polarized macrophages, and the inability to directly observe the interaction between neurons and macrophages from the same individuals. CONCLUSIONS: Our co-culture system revealed the non-cell autonomous adverse effects of GM-CSF MΦ in individuals with ASD on neurons, mediated by interleukin-1α and TNF-α. These results may support the immune dysfunction hypothesis of ASD, providing new insights into its pathology.


Assuntos
Transtorno do Espectro Autista , Citocinas , Feminino , Masculino , Humanos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Leucócitos Mononucleares/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-1alfa/farmacologia , Transtorno do Espectro Autista/metabolismo , Células Cultivadas , Sexismo , Macrófagos/metabolismo , Granulócitos/metabolismo , Dendritos/metabolismo
2.
Neurol Sci ; 44(12): 4511-4516, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37615876

RESUMO

BACKGROUND: Amyotrophic lateral sclerosis/Parkinsonism-dementia complex in Kii peninsula, Japan (Kii ALS/PDC), is an endemic neurodegenerative disease whose causes and pathogenesis remain unknown. However, astrocytes in autopsied cases of Kii ALS/PDC show characteristic lesions. In addition, relationships between extracellular vesicles (EVs) and neurodegenerative diseases are increasingly apparent. Therefore, we focused on proteins in EVs derived from Kii ALS/PDC astrocytes in the present study. METHODS: Induced pluripotent stem cells (iPSCs) derived from three healthy controls (HCs) and three patients with Kii ALS/PDC were differentiated into astrocytes. EVs in the culture medium of astrocytes were collected and subjected to quantitative proteome analysis. RESULTS: Our proteome analysis reveals that EV-containing proteins derived from astrocytes of patients with Kii ALS/PDC show distinctive patterns compared with those of HCs. Moreover, EVs derived from Kii ALS/PDC astrocytes display increased proteins related to proteostasis and decreased proteins related to anti-inflammation. DISCUSSION: Proteins contained in EVs from astrocytes unveil protective support to neurons and may reflect the molecular pathomechanism of Kii ALS/PDC; accordingly, they may be potential biomarker candidates of Kii ALS/PDC.


Assuntos
Esclerose Amiotrófica Lateral , Vesículas Extracelulares , Células-Tronco Pluripotentes Induzidas , Doenças Neurodegenerativas , Humanos , Esclerose Amiotrófica Lateral/epidemiologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Astrócitos/patologia , Proteoma , Japão/epidemiologia , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patologia
3.
Dalton Trans ; 52(29): 10206-10212, 2023 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-37435934

RESUMO

Platinum(II) complexes with salophen ligands bearing carboxy substituents at different positions, [Pt{(COOH)n-salophen}] (n = 2 (1), 3 (2), 1 (3)), were synthesized and characterized by acquiring UV-vis and luminescence spectra. These complexes exhibited systematic variations in absorption spectra depending on the number of carboxy groups, and this effect was attributed to metal-ligand charge transfer with support from density functional theory calculations. The luminescence properties of these complexes were also correlated with structural differences. Complexes 1-3 showed systematic spectral changes by addition of organic acid and base, respectively. This is based on the protonation/deprotonation of the carboxy substituents. Furthermore, aggregation-induced spectra change was investigated in DMSO-H2O mixtures with various proportions of water. Peak shifts in the range of 95 to 105 nm occurred in the absorption spectra in conjunction with pH changes. These variations resulted from molecular aggregation and diffusion associated with protonation/deprotonation of the carboxy groups. Variations in luminescence emission intensity and peak shifts were also observed. This work provides new insights into the correlations between the optical properties of carboxy-appended molecular complexes and pH changes and will assist in the future design of pH sensing devices based on molecular metal complexes.

4.
Int J Mol Sci ; 24(8)2023 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-37108151

RESUMO

Amyotrophic lateral sclerosis (ALS) is a major life-threatening disease caused by motor neuron degeneration. More effective treatments through drug discovery are urgently needed. Here, we established an effective high-throughput screening system using induced pluripotent stem cells (iPSCs). Using a Tet-On-dependent transcription factor expression system carried on the PiggyBac vector, motor neurons were efficiently and rapidly generated from iPSCs by a single-step induction method. Induced iPSC transcripts displayed characteristics similar to those of spinal cord neurons. iPSC-generated motor neurons carried a mutation in fused in sarcoma (FUS) and superoxide dismutase 1 (SOD1) genes and had abnormal protein accumulation corresponding to each mutation. Calcium imaging and multiple electrode array (MEA) recordings demonstrated that ALS neurons were abnormally hyperexcitable. Noticeably, protein accumulation and hyperexcitability were ameliorated by treatment with rapamycin (mTOR inhibitor) and retigabine (Kv7 channel activator), respectively. Furthermore, rapamycin suppressed ALS neuronal death and hyperexcitability, suggesting that protein aggregate clearance through the activation of autophagy effectively normalized activity and improved neuronal survival. Our culture system reproduced several ALS phenotypes, including protein accumulation, hyperexcitability, and neuronal death. This rapid and robust phenotypic screening system will likely facilitate the discovery of novel ALS therapeutics and stratified and personalized medicine for sporadic motor neuron diseases.


Assuntos
Esclerose Amiotrófica Lateral , Células-Tronco Pluripotentes Induzidas , Humanos , Esclerose Amiotrófica Lateral/metabolismo , Neurônios Motores/metabolismo , Superóxido Dismutase-1/metabolismo , Mutação , Fenótipo , Superóxido Dismutase/metabolismo
5.
Inflamm Regen ; 43(1): 23, 2023 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-37009886

RESUMO

Cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL) is an inherited cerebral small vessel disease (CSVD) caused by biallelic mutations in the high-temperature requirement serine peptidase A1 (HTRA1) gene. Even heterozygous mutations in HTRA1 are recently revealed to cause cardinal clinical features of CSVD. Here, we report the first establishment of a human induced pluripotent stem cell (hiPSC) line from a patient with heterozygous HTRA1-related CSVD. Peripheral blood mononuclear cells (PBMCs) were reprogrammed by the transfection of episomal vectors encoding human OCT3/4 (POU5F1), SOX2, KLF4, L-MYC, LIN28, and a murine dominant-negative mutant of p53 (mp53DD). The established iPSCs had normal morphology as human pluripotent stem cells and normal karyotype (46XX). Moreover, we found that the HTRA1 missense mutation (c.905G>A, p.R302Q) was heterozygous. These iPSCs expressed pluripotency-related markers and had the potential to differentiate into all three germ layers in vitro. HTRA1 and the supposed disease-associated gene NOG were differentially expressed in the patient iPSCs at mRNA levels compared to those of control lines. The iPSC line would facilitate in vitro research for understanding the cellular pathomechanisms caused by the HTRA1 mutation including its dominant-negative effect.

6.
Methods Mol Biol ; 2509: 143-153, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35796962

RESUMO

Transposable elements (TEs) constitute a large proportion of the genome in multiple organisms. Therefore, anti-transposable element machineries are essential to maintain genomic integrity. PIWI-interacting RNAs (piRNAs) are a major force to repress TEs in Drosophila ovaries. Ovarian somatic cells (OSC), in which nuclear piRNA regulation is functional, have been used for research on piRNA pathway as a cell culture system to elucidate the molecular mechanisms underlying the piRNA pathway. Analysis of piRNA pathway using a reporter system to monitor the gene regulation or overexpression of specific genes would be a powerful approach. Here, we present the technical protocol to establish stable cell lines using the piggyBac system, adopted for OSCs. This easy, consistent, and timesaving protocol may accelerate research on the piRNA pathway.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Linhagem Celular , Elementos de DNA Transponíveis/genética , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Feminino , Ovário/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
7.
Regen Ther ; 20: 165-186, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35620640

RESUMO

Introduction: Efficient induction of the otic placode, the developmental origin of the inner ear from human pluripotent stem cells (hPSCs), provides a robust platform for otic development and sensorineural hearing loss modelling. Nevertheless, there remains a limited capacity of otic lineage specification from hPSCs by stepwise differentiation methods, since the critical factors for successful otic cell differentiation have not been thoroughly investigated. In this study, we developed a novel differentiation system involving the use of a three-dimensional (3D) floating culture with signalling factors for generating otic cell lineages via stepwise differentiation of hPSCs. Methods: We differentiated hPSCs into preplacodal cells under a two-dimensional (2D) monolayer culture. Then, we transferred the induced preplacodal cells into a 3D floating culture under the control of the fibroblast growth factor (FGF), bone morphogenetic protein (BMP), retinoic acid (RA) and WNT signalling pathways. We evaluated the characteristics of the induced cells using immunocytochemistry, quantitative PCR (qPCR), population averaging, and single-cell RNA-seq (RNA-seq) analysis. We further investigated the methods for differentiating otic progenitors towards hair cells by overexpression of defined transcription factors. Results: We demonstrated that hPSC-derived preplacodal cells acquired the potential to differentiate into posterior placodal cells in 3D floating culture with FGF2 and RA. Subsequent activation of WNT signalling induced otic placodal cell formation. By single-cell RNA-seq (scRNA-seq) analysis, we identified multiple clusters of otic placode- and otocyst marker-positive cells in the induced spheres. Moreover, the induced otic cells showed the potential to generate hair cell-like cells by overexpression of the transcription factors ATOH1, POU4F3 and GFI1. Conclusions: We demonstrated the critical role of FGF2, RA and WNT signalling in a 3D environment for the in vitro differentiation of otic lineage cells from hPSCs. The induced otic cells had the capacity to differentiate into inner ear hair cells with stereociliary bundles and tip link-like structures. The protocol will be useful for in vitro disease modelling of sensorineural hearing loss and human inner ear development and thus contribute to drug screening and stem cell-based regenerative medicine.

8.
Cell Rep ; 37(8): 110019, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34818559

RESUMO

In cell transplantation therapy for spinal cord injury (SCI), grafted human induced pluripotent stem cell-derived neural stem/progenitor cells (hiPSC-NS/PCs) mainly differentiate into neurons, forming synapses in a process similar to neurodevelopment. In the developing nervous system, the activity of immature neurons has an important role in constructing and maintaining new synapses. Thus, we investigate how enhancing the activity of transplanted hiPSC-NS/PCs affects both the transplanted cells themselves and the host tissue. We find that chemogenetic stimulation of hiPSC-derived neural cells enhances cell activity and neuron-to-neuron interactions in vitro. In a rodent model of SCI, consecutive and selective chemogenetic stimulation of transplanted hiPSC-NS/PCs also enhances the expression of synapse-related genes and proteins in surrounding host tissues and prevents atrophy of the injured spinal cord, thereby improving locomotor function. These findings provide a strategy for enhancing activity within the graft to improve the efficacy of cell transplantation therapy for SCI.


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Locomoção/fisiologia , Traumatismos da Medula Espinal/terapia , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Células Cultivadas , Modelos Animais de Doenças , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Camundongos , Camundongos SCID , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/fisiologia , Células-Tronco Neurais/transplante , Neurônios/metabolismo , Recuperação de Função Fisiológica , Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/fisiopatologia , Transplante de Células-Tronco/métodos
9.
Stem Cell Res ; 56: 102549, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34598113

RESUMO

The defective and persistent Sendai virus (SeVdp) vector system allows efficient generation of transgene-free induced pluripotent stem cells (iPSCs) from human somatic cells. By leveraging the system, here we report the generation of an iPSC line from somatic fibroblasts of a healthy control donner (female), named KEIOi002-A (also named YG-iPS). The control iPSC line would be a useful resource for stem cell research and regenerative medicine.


Assuntos
Células-Tronco Pluripotentes Induzidas , Diferenciação Celular , Reprogramação Celular , Feminino , Fibroblastos , Vetores Genéticos/genética , Humanos , Vírus Sendai/genética , Transgenes
10.
Stem Cell Res ; 53: 102380, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34088009

RESUMO

Epstein-Barr virus (EBV)-based episomal vector system enables persistent transgene expression, which is advantageous for efficient derivation of transgene-free induced pluripotent stem cells (iPSCs) without viral transduction. Here, we report establishment of an iPSC line from somatic fibroblasts of a neonatal common marmoset monkey (marmoset; Callithrix jacchus) using an all-in-one episomal vector that we newly developed. The established iPSC line, named NM-iPS, showed standard characteristics of pluripotency such as pluripotency-related marker expression, three germ layer differentiation, and normal karyotype (2n = 46). The novel iPSC line would be a useful resource for stem cell research using non-human primates.


Assuntos
Infecções por Vírus Epstein-Barr , Células-Tronco Pluripotentes Induzidas , Animais , Callithrix , Diferenciação Celular , Fibroblastos , Herpesvirus Humano 4
11.
Stem Cell Reports ; 16(6): 1527-1541, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34048688

RESUMO

Amyotrophic lateral sclerosis (ALS) is an adult-onset incurable motor neuron (MN) disease. The reasons for selective MN vulnerability in ALS are unknown. Axonal pathology is among the earliest signs of ALS. We searched for novel modulatory genes in human MN axon shortening affected by TARDBP mutations. In transcriptome analysis of RNA present in the axon compartment of human-derived induced pluripotent stem cell (iPSC)-derived MNs, PHOX2B (paired-like homeobox protein 2B) showed lower expression in TARDBP mutant axons, which was consistent with axon qPCR and in situ hybridization. PHOX2B mRNA stability was reduced in TARDBP mutant MNs. Furthermore, PHOX2B knockdown reduced neurite length in human MNs. Finally, phox2b knockdown in zebrafish induced short spinal axons and impaired escape response. PHOX2B is known to be highly express in other types of neurons maintained after ALS progression. Collectively, TARDBP mutations induced loss of axonal resilience, which is an important ALS-related phenotype mediated by PHOX2B downregulation.


Assuntos
Esclerose Amiotrófica Lateral/metabolismo , Axônios/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios Motores/metabolismo , Fatores de Transcrição/metabolismo , Peixe-Zebra/metabolismo , Esclerose Amiotrófica Lateral/genética , Animais , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes/métodos , Proteínas de Homeodomínio/genética , Humanos , Mutação , Fenótipo , Fatores de Transcrição/genética , Transcriptoma
12.
Hum Mol Genet ; 30(14): 1337-1348, 2021 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-33961044

RESUMO

Syntaxin-binding protein 1 (STXBP1; also called MUNC18-1), encoded by STXBP1, is an essential component of the molecular machinery that controls synaptic vesicle docking and fusion. De novo pathogenic variants of STXBP1 cause a complex set of neurological disturbances, namely STXBP1 encephalopathy (STXBP1-E) that includes epilepsy, neurodevelopmental disorders and neurodegeneration. Several animal studies have suggested the contribution of GABAergic dysfunction in STXBP1-E pathogenesis. However, the pathophysiological changes in GABAergic neurons of these patients are still poorly understood. Here, we exclusively generated GABAergic neurons from STXBP1-E patient-derived induced pluripotent stem cells (iPSCs) by transient expression of the transcription factors ASCL1 and DLX2. We also generated CRISPR/Cas9-edited isogenic iPSC-derived GABAergic (iPSC GABA) neurons as controls. We demonstrated that the reduction in STXBP1 protein levels in patient-derived iPSC GABA neurons was slight (approximately 20%) compared to the control neurons, despite a 50% reduction in STXBP1 mRNA levels. Using a microelectrode array-based assay, we found that patient-derived iPSC GABA neurons exhibited dysfunctional maturation with reduced numbers of spontaneous spikes and bursts. These findings reinforce the idea that GABAergic dysfunction is a crucial contributor to STXBP1-E pathogenesis. Moreover, gene expression analysis revealed specific dysregulation of genes previously implicated in epilepsy, neurodevelopment and neurodegeneration in patient-derived iPSC GABA neurons, namely KCNH1, KCNH5, CNN3, RASGRF1, SEMA3A, SIAH3 and INPP5F. Thus, our study provides new insights for understanding the biological processes underlying the widespread neuropathological features of STXBP1-E.


Assuntos
Encefalopatias , Células-Tronco Pluripotentes Induzidas , Animais , Encefalopatias/genética , Encefalopatias/metabolismo , Neurônios GABAérgicos/metabolismo , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas Munc18/genética , Proteínas Munc18/metabolismo
13.
Stem Cell Reports ; 16(4): 754-770, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33798453

RESUMO

Induced pluripotent stem cells (iPSCs) are capable of providing an unlimited source of cells from all three germ layers and germ cells. The derivation and usage of iPSCs from various animal models may facilitate stem cell-based therapy, gene-modified animal production, and evolutionary studies assessing interspecies differences. However, there is a lack of species-wide methods for deriving iPSCs, in particular by means of non-viral and non-transgene-integrating (NTI) approaches. Here, we demonstrate the iPSC derivation from somatic fibroblasts of multiple mammalian species from three different taxonomic orders, including the common marmoset (Callithrix jacchus) in Primates, the dog (Canis lupus familiaris) in Carnivora, and the pig (Sus scrofa) in Cetartiodactyla, by combinatorial usage of chemical compounds and NTI episomal vectors. Interestingly, the fibroblasts temporarily acquired a neural stem cell-like state during the reprogramming. Collectively, our method, robustly applicable to various species, holds a great potential for facilitating stem cell-based research using various animals in Mammalia.


Assuntos
Fibroblastos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Mamíferos/metabolismo , Transgenes , Animais , Callithrix , Cães , Perfilação da Expressão Gênica , Vetores Genéticos/metabolismo , Camadas Germinativas/metabolismo , Células-Tronco Neurais/metabolismo , Plasmídeos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA-Seq , Especificidade da Espécie , Suínos , Vírus
14.
Neurobiol Dis ; 155: 105364, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33857636

RESUMO

Fused in sarcoma/translated in liposarcoma (FUS) is a causative gene of amyotrophic lateral sclerosis (ALS). Mutated FUS causes accumulation of DNA damage and cytosolic stress granule (SG) formation, thereby motor neuron (MN) death. However, key molecular aetiology remains unclear. Here, we applied a novel platform technology, iBRN, "Non- biased" Bayesian gene regulatory network analysis based on induced pluripotent stem cell (iPSC)-derived cell model, to elucidate the molecular aetiology using transcriptome of iPSC-derived MNs harboring FUSH517D. iBRN revealed "hub molecules", which strongly influenced transcriptome network, such as miR-125b-5p-TIMELESS axis and PRKDC for the molecular aetiology. Next, we confirmed miR-125b-5p-TIMELESS axis in FUSH517D MNs such that miR-125b-5p regulated several DNA repair-related genes including TIMELESS. In addition, we validated both introduction of miR-125b-5p and knocking down of TIMELESS caused DNA damage in the cell culture model. Furthermore, PRKDC was strongly associated with FUS mis-localization into SGs by DNA damage under impaired DNA-PK activity. Collectively, our iBRN strategy provides the first compelling evidence to elucidate molecular aetiology in neurodegenerative diseases.


Assuntos
Esclerose Amiotrófica Lateral/genética , Redes Reguladoras de Genes/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , MicroRNAs/genética , Proteína FUS de Ligação a RNA/genética , Esclerose Amiotrófica Lateral/metabolismo , Teorema de Bayes , Linhagem Celular Tumoral , Dano ao DNA/fisiologia , Técnicas de Inativação de Genes/métodos , Humanos , MicroRNAs/biossíntese , Proteína FUS de Ligação a RNA/biossíntese
15.
Int J Mol Sci ; 22(8)2021 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-33924373

RESUMO

A common pathological hallmark of several neurodegenerative diseases, including amyotrophic lateral sclerosis, is cytoplasmic mislocalization and aggregation of nuclear RNA-binding protein TDP-43. Perry disease, which displays inherited atypical parkinsonism, is a type of TDP-43 proteinopathy. The causative gene DCTN1 encodes the largest subunit of the dynactin complex. Dynactin associates with the microtubule-based motor cytoplasmic dynein and is required for dynein-mediated long-distance retrograde transport. Perry disease-linked missense mutations (e.g., p.G71A) reside within the CAP-Gly domain and impair the microtubule-binding abilities of DCTN1. However, molecular mechanisms by which such DCTN1 mutations cause TDP-43 proteinopathy remain unclear. We found that DCTN1 bound to TDP-43. Biochemical analysis using a panel of truncated mutants revealed that the DCTN1 CAP-Gly-basic supradomain, dynactin domain, and C-terminal region interacted with TDP-43, preferentially through its C-terminal region. Remarkably, the p.G71A mutation affected the TDP-43-interacting ability of DCTN1. Overexpression of DCTN1G71A, the dynactin-domain fragment, or C-terminal fragment, but not the CAP-Gly-basic fragment, induced cytoplasmic mislocalization and aggregation of TDP-43, suggesting functional modularity among TDP-43-interacting domains of DCTN1. We thus identified DCTN1 as a new player in TDP-43 cytoplasmic-nuclear transport, and showed that dysregulation of DCTN1-TDP-43 interactions triggers mislocalization and aggregation of TDP-43, thus providing insights into the pathological mechanisms of Perry disease and other TDP-43 proteinopathies.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Complexo Dinactina/metabolismo , Agregados Proteicos , Sequência de Aminoácidos , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Complexo Dinactina/química , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Neurônios/metabolismo , Sinais de Localização Nuclear/metabolismo , Mutação Puntual/genética , Ligação Proteica , Frações Subcelulares/metabolismo
16.
Neurosci Lett ; 746: 135676, 2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33516803

RESUMO

Human induced pluripotent stem cells (iPSCs) have great potential to elucidate the molecular pathogenesis of neurological/psychiatric diseases. In particular, neurological/psychiatric diseases often display brain region-specific symptoms, and the technology for generating region-specific neural cells from iPSCs has been established for detailed modeling of neurological/psychiatric disease phenotypes in vitro. On the other hand, recent advances in culturing human iPSCs without feeder cells have enabled highly efficient and reproducible neural induction. However, conventional regional control technologies have mainly been developed based on on-feeder iPSCs, and these methods are difficult to apply to feeder-free (ff) iPSC cultures. In this study, we established a novel culture system to generate region-specific neural cells from human ff-iPSCs. This system is the best optimized approach for feeder-free iPSC culture and generates specific neuronal subtypes with high purity and functionality, including forebrain cortical neurons, forebrain interneurons, midbrain dopaminergic neurons, and spinal motor neurons. In addition, the temporal patterning of cortical neuron layer specification in the forebrain was reproduced in our culture system, which enables the generation of layer-specific cortical neurons. Neuronal activity was demonstrated in the present culture system by using multiple electrode array and calcium imaging. Collectively, our ff-iPSC-based culture system would provide a desirable platform for modeling various types of neurological/psychiatric disease phenotypes.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Neurônios/fisiologia , Humanos , Imagem Molecular/métodos
17.
Cells ; 9(12)2020 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-33322219

RESUMO

Induced pluripotent stem cell (iPSC)-based disease modeling has a great potential for uncovering the mechanisms of pathogenesis, especially in the case of neurodegenerative diseases where disease-susceptible cells can usually not be obtained from patients. So far, the iPSC-based modeling of neurodegenerative diseases has mainly focused on neurons because the protocols for generating astrocytes from iPSCs have not been fully established. The growing evidence of astrocytes' contribution to neurodegenerative diseases has underscored the lack of iPSC-derived astrocyte models. In the present study, we established a protocol to efficiently generate iPSC-derived astrocytes (iPasts), which were further characterized by RNA and protein expression profiles as well as functional assays. iPasts exhibited calcium dynamics and glutamate uptake activity comparable to human primary astrocytes. Moreover, when co-cultured with neurons, iPasts enhanced neuronal synaptic maturation. Our protocol can be used for modeling astrocyte-related disease phenotypes in vitro and further exploring the contribution of astrocytes to neurodegenerative diseases.


Assuntos
Astrócitos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Astrócitos/citologia , Astrócitos/patologia , Cálcio/metabolismo , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Técnicas de Cocultura , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Ácido Glutâmico/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Microscopia de Fluorescência , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/citologia , Neurônios/metabolismo , Neurônios/patologia , Análise de Componente Principal
18.
Stem Cell Res ; 49: 102073, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33181472

RESUMO

Leucine-rich repeat kinase 2 (LRRK2) is the causal gene of the autosomal dominant hereditary form of Parkinson's disease (PD), PARK8. We have previously reported that induced pluripotent stem cells (iPSCs) from a PARK8 patient with I2020T LRRK2 mutation replicated to some extent the pathologic phenotype evident in the brain of PD patients. In the present study, we generated gene-corrected iPSCs line, KEIUi001-A, using TALEN-mediated genome editing. KEIUi001-A retained a normal karyotype and pluripotency, i.e. the capacity to differentiate into cell types of the three germ layers. This iPSCs will be valuable for clarifying various aspects of LRRK2-related pathology.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doença de Parkinson , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Mutação , Doença de Parkinson/genética , Fenótipo
19.
Biochem Biophys Res Commun ; 529(3): 615-621, 2020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-32736682

RESUMO

Suppressor of cancer cell invasion (SCAI) is a suppressor of myocardin-related transcription factor (MRTF)-mediated transcription and cancer cell invasion. However, roles of SCAI in the brain and neuronal cells are not fully resolved. In this study, we initially investigated the distribution of Scai mRNA in the developing rat brain and in neurons. We found that, although Scai mRNA levels decreased during brain development, it was highly expressed in several brain regions and in neurons but not astrocytes. Subsequently, in addition to Scai variant 1, we identified novel rat Scai variants 2 and 3 and characterized their functions in Neuro-2a cells. The novel Scai variants 2 and 3 contain unique exons that possess stop codons and therefore encode shorter proteins compared with the full-length Scai variant 1. SCAI variants 2 and 3 possess a nuclear localization signal, but do not have an MRTF-binding site. Immunostaining of green fluorescent protein (GFP)-tagged SCAI variants revealed a nuclear localization of variant 1, whereas localization of variants 2 and 3 was throughout the cytoplasm and nucleus, suggesting that other nuclear localization signals, which act in Neuro-2a cells, exist in SCAI. All three SCAI variants suppressed the neuron-like morphological change of Neuro-2a cells induced by a Rho effector, constitutively active mDia; however, the suppressive effects of variants 2 and 3 were weaker than that of full-length SCAI variant 1, indicating that the SCAI-mediated change toward a neuronal morphology appeared to be consistent with their nuclear localization. These findings indicate that generation of multiple SCAI splice variants fines-tune neuronal morphology.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Neurônios/metabolismo , Splicing de RNA , Fatores de Transcrição/genética , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Células NIH 3T3 , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratos Sprague-Dawley , Fatores de Transcrição/metabolismo
20.
Mol Cell Neurosci ; 108: 103535, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32758699

RESUMO

Epilepsy is among the most common neurological disorders, affecting approximately 50 million people worldwide. Importantly, epilepsy is genetically and etiologically heterogenous, but several epilepsy types exhibit similar clinical presentations. Epilepsy-associated genes are being identified. However, the molecular pathomechanisms remain largely unknown. Approximately one-third of epilepsy is refractory to multiple conventional anti-epileptic drugs (AEDs). Induced pluripotent stem cells (iPSCs) provide an excellent tool to study the pathomechanisms underlying epilepsy and to develop novel treatments. Indeed, disease-specific iPSCs have been established for several genetic epilepsies. In particular, the molecular mechanisms underlying certain developmental and epileptic encephalopathies, such as Dravet syndrome, have been revealed. Modeling epilepsy with iPSCs enables new drug development based on the elucidated pathomechanisms. This can also be used to evaluate conventional AEDs and drug repurposing. Furthermore, transplanting neuronal cells derived from iPSCs into the brain has great potential to treat refractory epilepsies. Recent advances in iPSC technology have enabled the generation of neuronal organoids, or "mini brains." These organoids demonstrate electrophysiological activities similar to those of the brain and have the potential for extensive epilepsy research opportunities. Thus, the application of iPSCs in epilepsy provides insight into novel treatments based on the molecular pathomechanisms of epilepsy. In this review, we comprehensively discuss the studies conducted on iPSCs established for genetic epilepsy or epilepsies without major structural dysmorphic features.


Assuntos
Anticonvulsivantes/farmacologia , Epilepsia/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Avaliação Pré-Clínica de Medicamentos/métodos , Epilepsia/genética , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...