Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 971
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Neurosci ; 44: 27-48, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34236888

ABSTRACT

The common marmoset (Callithrix jacchus), a small New World primate, is receiving substantial attention in the neuroscience and biomedical science fields because its anatomical features, functional and behavioral characteristics, and reproductive features and its amenability to available genetic modification technologies make it an attractive experimental subject. In this review, I outline the progress of marmoset neuroscience research and summarize both the current status (opportunities and limitations) of and the future perspectives on the application of marmosets in neuroscience and disease modeling.


Subject(s)
Callithrix , Neurosciences , Animals , Neurobiology , Reproduction
2.
EMBO J ; 42(16): e111133, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37431790

ABSTRACT

Naked mole-rats (NMRs) have exceptional longevity and are resistant to age-related physiological decline and diseases. Given the role of cellular senescence in aging, we postulated that NMRs possess unidentified species-specific mechanisms to prevent senescent cell accumulation. Here, we show that upon induction of cellular senescence, NMR fibroblasts underwent delayed and progressive cell death that required activation of the INK4a-retinoblastoma protein (RB) pathway (termed "INK4a-RB cell death"), a phenomenon not observed in mouse fibroblasts. Naked mole-rat fibroblasts uniquely accumulated serotonin and were inherently vulnerable to hydrogen peroxide (H2 O2 ). After activation of the INK4a-RB pathway, NMR fibroblasts increased monoamine oxidase levels, leading to serotonin oxidization and H2 O2 production, which resulted in increased intracellular oxidative damage and cell death activation. In the NMR lung, induction of cellular senescence caused delayed, progressive cell death mediated by monoamine oxidase activation, thereby preventing senescent cell accumulation, consistent with in vitro results. The present findings indicate that INK4a-RB cell death likely functions as a natural senolytic mechanism in NMRs, providing an evolutionary rationale for senescent cell removal as a strategy to resist aging.


Subject(s)
Cellular Senescence , Serotonin , Animals , Mice , Serotonin/metabolism , Cellular Senescence/physiology , Aging/metabolism , Cell Death , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Mole Rats/metabolism
3.
PLoS Biol ; 21(6): e3002158, 2023 06.
Article in English | MEDLINE | ID: mdl-37384809

ABSTRACT

The primate brain has unique anatomical characteristics, which translate into advanced cognitive, sensory, and motor abilities. Thus, it is important that we gain insight on its structure to provide a solid basis for models that will clarify function. Here, we report on the implementation and features of the Brain/MINDS Marmoset Connectivity Resource (BMCR), a new open-access platform that provides access to high-resolution anterograde neuronal tracer data in the marmoset brain, integrated to retrograde tracer and tractography data. Unlike other existing image explorers, the BMCR allows visualization of data from different individuals and modalities in a common reference space. This feature, allied to an unprecedented high resolution, enables analyses of features such as reciprocity, directionality, and spatial segregation of connections. The present release of the BMCR focuses on the prefrontal cortex (PFC), a uniquely developed region of the primate brain that is linked to advanced cognition, including the results of 52 anterograde and 164 retrograde tracer injections in the cortex of the marmoset. Moreover, the inclusion of tractography data from diffusion MRI allows systematic analyses of this noninvasive modality against gold-standard cellular connectivity data, enabling detection of false positives and negatives, which provide a basis for future development of tractography. This paper introduces the BMCR image preprocessing pipeline and resources, which include new tools for exploring and reviewing the data.


Subject(s)
Brain , Callithrix , Animals , Brain/diagnostic imaging , Brain/physiology , Brain Mapping/methods , Prefrontal Cortex/diagnostic imaging , Diffusion Magnetic Resonance Imaging , Neural Pathways
4.
Nature ; 585(7826): 591-596, 2020 09.
Article in English | MEDLINE | ID: mdl-32526765

ABSTRACT

Recent clinical and experimental evidence has evoked the concept of the gut-brain axis to explain mutual interactions between the central nervous system and gut microbiota that are closely associated with the bidirectional effects of inflammatory bowel disease and central nervous system disorders1-4. Despite recent advances in our understanding of neuroimmune interactions, it remains unclear how the gut and brain communicate to maintain gut immune homeostasis, including in the induction and maintenance of peripheral regulatory T cells (pTreg cells), and what environmental cues prompt the host to protect itself from development of inflammatory bowel diseases. Here we report a liver-brain-gut neural arc that ensures the proper differentiation and maintenance of pTreg cells in the gut. The hepatic vagal sensory afferent nerves are responsible for indirectly sensing the gut microenvironment and relaying the sensory inputs to the nucleus tractus solitarius of the brainstem, and ultimately to the vagal parasympathetic nerves and enteric neurons. Surgical and chemical perturbation of the vagal sensory afferents at the hepatic afferent level reduced the abundance of colonic pTreg cells; this was attributed to decreased aldehyde dehydrogenase (ALDH) expression and retinoic acid synthesis by intestinal antigen-presenting cells. Activation of muscarinic acetylcholine receptors directly induced ALDH gene expression in both human and mouse colonic antigen-presenting cells, whereas genetic ablation of these receptors abolished the stimulation of antigen-presenting cells in vitro. Disruption of left vagal sensory afferents from the liver to the brainstem in mouse models of colitis reduced the colonic pTreg cell pool, resulting in increased susceptibility to colitis. These results demonstrate that the novel vago-vagal liver-brain-gut reflex arc controls the number of pTreg cells and maintains gut homeostasis. Intervention in this autonomic feedback feedforward system could help in the development of therapeutic strategies to treat or prevent immunological disorders of the gut.


Subject(s)
Brain/cytology , Intestines/cytology , Intestines/innervation , Liver/cytology , Liver/innervation , Neurons/physiology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Afferent Pathways , Animals , Antigen-Presenting Cells/immunology , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Homeostasis , Humans , Intestines/immunology , Male , Mice , Rats , Receptors, Muscarinic/metabolism , Spleen/cytology , Spleen/immunology , Vagus Nerve/physiology
5.
Genes Dev ; 32(2): 165-180, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29440260

ABSTRACT

Multiple congenital disorders often present complex phenotypes, but how the mutation of individual genetic factors can lead to multiple defects remains poorly understood. In the present study, we used human neuroepithelial (NE) cells and CHARGE patient-derived cells as an in vitro model system to identify the function of chromodomain helicase DNA-binding 7 (CHD7) in NE-neural crest bifurcation, thus revealing an etiological link between the central nervous system (CNS) and craniofacial anomalies observed in CHARGE syndrome. We found that CHD7 is required for epigenetic activation of superenhancers and CNS-specific enhancers, which support the maintenance of the NE and CNS lineage identities. Furthermore, we found that BRN2 and SOX21 are downstream effectors of CHD7, which shapes cellular identities by enhancing a CNS-specific cellular program and indirectly repressing non-CNS-specific cellular programs. Based on our results, CHD7, through its interactions with superenhancer elements, acts as a regulatory hub in the orchestration of the spatiotemporal dynamics of transcription factors to regulate NE and CNS lineage identities.


Subject(s)
DNA Helicases/physiology , DNA-Binding Proteins/physiology , Epigenesis, Genetic , Neural Stem Cells/metabolism , Neuroepithelial Cells/metabolism , CHARGE Syndrome/genetics , Cell Line , Cell Lineage/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Haploinsufficiency , Humans , Neural Crest/metabolism , Transcription, Genetic
6.
J Neurosci ; 44(24)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38649269

ABSTRACT

Genetic variants in the apolipoprotein E (APOE) gene affect the onset and progression of Alzheimer's disease (AD). The APOE Christchurch (APOE Ch) variant has been identified as the most prominent candidate for preventing the onset and progression of AD. In this study, we generated isogenic APOE3Ch/3Ch human-induced pluripotent stem cells (iPSCs) from APOE3/3 healthy control female iPSCs and induced them into astrocytes. RNA expression analysis revealed the inherent resilience of APOE3Ch/3Ch astrocytes to induce a reactive state in response to inflammatory cytokines. Moreover, cytokine treatment changed astrocytic morphology with more complexity in APOE3/3 astrocytes, but not in APOE3Ch/3Ch astrocytes, indicating resilience of the rare variant to a reactive state. Interestingly, we observed robust morphological alterations containing more intricate processes when cocultured with iPSC-derived cortical neurons, in which APOE3Ch/3Ch astrocytes reduced complexity compared with APOE3/3 astrocytes. To assess the impacts of tau propagation effects, we next developed a sophisticated and sensitive assay utilizing cortical neurons derived from human iPSCs, previously generated from donors of both sexes. We showed that APOE3Ch/3Ch astrocytes effectively mitigated tau propagation within iPSC-derived neurons. This study provides important experimental evidence of the characteristic functions exhibited by APOE3Ch/3Ch astrocytes, thereby offering valuable insights for the advancement of novel clinical interventions in AD research.


Subject(s)
Astrocytes , Induced Pluripotent Stem Cells , tau Proteins , Astrocytes/metabolism , Humans , tau Proteins/metabolism , tau Proteins/genetics , Female , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Apolipoprotein E3/genetics , Neurons/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cells, Cultured , Coculture Techniques
7.
Genes Dev ; 31(18): 1910-1925, 2017 09 15.
Article in English | MEDLINE | ID: mdl-29021239

ABSTRACT

Cell type-specific transcriptomes are enabled by the action of multiple regulators, which are frequently expressed within restricted tissue regions. In the present study, we identify one such regulator, Quaking 5 (Qki5), as an RNA-binding protein (RNABP) that is expressed in early embryonic neural stem cells and subsequently down-regulated during neurogenesis. mRNA sequencing analysis in neural stem cell culture indicates that Qki proteins play supporting roles in the neural stem cell transcriptome and various forms of mRNA processing that may result from regionally restricted expression and subcellular localization. Also, our in utero electroporation gain-of-function study suggests that the nuclear-type Qki isoform Qki5 supports the neural stem cell state. We next performed in vivo transcriptome-wide protein-RNA interaction mapping to search for direct targets of Qki5 and elucidate how Qki5 regulates neural stem cell function. Combined with our transcriptome analysis, this mapping analysis yielded a bona fide map of Qki5-RNA interaction at single-nucleotide resolution, the identification of 892 Qki5 direct target genes, and an accurate Qki5-dependent alternative splicing rule in the developing brain. Last, our target gene list provides the first compelling evidence that Qki5 is associated with specific biological events; namely, cell-cell adhesion. This prediction was confirmed by histological analysis of mice in which Qki proteins were genetically ablated, which revealed disruption of the apical surface of the lateral wall in the developing brain. These data collectively indicate that Qki5 regulates communication between neural stem cells by mediating numerous RNA processing events and suggest new links between splicing regulation and neural stem cell states.


Subject(s)
Brain/embryology , Cell Adhesion/physiology , Mouse Embryonic Stem Cells/metabolism , Neural Stem Cells/metabolism , RNA Precursors/metabolism , RNA-Binding Proteins/metabolism , Alternative Splicing/physiology , Animals , Cell Communication , Down-Regulation , Gene Expression Profiling , Mice , Mice, Knockout , Neurogenesis/genetics , Neurogenesis/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Signal Transduction
8.
Genes Cells ; 28(7): 496-502, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37096945

ABSTRACT

Understanding the molecular genetic basis of animal magnet reception has been one of the big challenges in molecular biology. Recently it was discovered that the magnetic sense of Drosophila melanogaster is mediated by the ultraviolet (UV)-A/blue light photoreceptor cryptochrome (Cry). Here, using the fruit fly as a magnet-receptive model organism, we show that the magnetic field exposure (0.4-0.6 mT) extended lifespan under starvation, but not in cryptochrome mutant flies (cryb ). The magnetic field exposure increases motor function in wild type and neurodegenerative disease model flies. Furthermore, the magnetic field exposure improved sleep quality at night-time specific manner, but not in cryb . We also showed that repeated AC magnetic field exposure increased climbing activity in wild-type Drosophila, but not in cryb . The data suggests that magnetic field-dependent improvement of lifespan, sleep quality, and motor function is mediated through a cry-dependent pathway in Drosophila.


Subject(s)
Cryptochromes , Drosophila Proteins , Drosophila melanogaster , Magnetic Fields , Animals , Circadian Rhythm/physiology , Cryptochromes/genetics , Cryptochromes/metabolism , Drosophila melanogaster/metabolism , Drosophila Proteins/metabolism , Light , Longevity , Neurodegenerative Diseases , Sleep Quality
9.
Genes Cells ; 28(1): 29-41, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36401755

ABSTRACT

The coronavirus disease 2019 (COVID-19) epidemic remains worldwide. The usefulness of the intranasal vaccine and boost immunization against severe acute respiratory syndrome-related coronavirus (SARS-CoV-2) has recently received much attention. We developed an intranasal SARS-CoV-2 vaccine by loading the receptor binding domain of the S protein (S-RBD) of SARS-CoV-2 as an antigen into an F-deficient Sendai virus vector. After the S-RBD-Fd antigen with trimer formation ability was intranasally administered to mice, S-RBD-specific IgM, IgG, IgA, and neutralizing antibody titers were increased in serum or bronchoalveolar lavage fluid for 12 weeks. Furthermore, in mice that received a booster dose at week 8, a marked increase in neutralizing antibodies in the serum and bronchoalveolar lavage fluid was observed at the final evaluation at week 12, which neutralized the pseudotyped lentivirus expressing the SARS-CoV-2 spike protein, indicating the usefulness of the Sendai virus-based SARS-CoV-2 intranasal vaccine.


Subject(s)
COVID-19 Vaccines , COVID-19 , Animals , Antibodies, Viral , COVID-19/prevention & control , Disease Models, Animal , SARS-CoV-2 , Sendai virus/genetics , Mice
10.
Genes Cells ; 28(2): 156-169, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36530170

ABSTRACT

Extended pluripotent stem cells (EPSCs) derived from mice and humans showed an enhanced potential for chimeric formation. By exploiting transcriptomic approaches, we assessed the differences in gene expression profile between extended EPSCs derived from mice and humans, and those newly derived from the common marmoset (marmoset; Callithrix jacchus). Although the marmoset EPSC-like cells displayed a unique colony morphology distinct from murine and human EPSCs, they displayed a pluripotent state akin to embryonic stem cells (ESCs), as confirmed by gene expression and immunocytochemical analyses of pluripotency markers and three-germ-layer differentiation assay. Importantly, the marmoset EPSC-like cells showed interspecies chimeric contribution to mouse embryos, such as E6.5 blastocysts in vitro and E6.5 epiblasts in vivo in mouse development. Also, we discovered that the perturbation of gene expression of the marmoset EPSC-like cells from the original ESCs resembled that of human EPSCs. Taken together, our multiple analyses evaluated the efficacy of the method for the derivation of marmoset EPSCs.


Subject(s)
Callithrix , Embryonic Stem Cells , Animals , Humans , Mice , Embryonic Stem Cells/metabolism , Cell Differentiation , Gene Expression Profiling , Transcriptome
11.
Acta Neuropathol ; 147(1): 84, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38750212

ABSTRACT

Amyotrophic Lateral Sclerosis/Parkinsonism-Dementia Complex (ALS/PDC), a rare and complex neurological disorder, is predominantly observed in the Western Pacific islands, including regions of Japan, Guam, and Papua. This enigmatic condition continues to capture medical attention due to affected patients displaying symptoms that parallel those seen in either classical amyotrophic lateral sclerosis (ALS) or Parkinson's disease (PD). Distinctly, postmortem examinations of the brains of affected individuals have shown the presence of α-synuclein aggregates and TDP-43, which are hallmarks of PD and classical ALS, respectively. These observations are further complicated by the detection of phosphorylated tau, accentuating the multifaceted proteinopathic nature of ALS/PDC. The etiological foundations of this disease remain undetermined, and genetic investigations have yet to provide conclusive answers. However, emerging evidence has implicated the contribution of astrocytes, pivotal cells for maintaining brain health, to neurodegenerative onset, and likely to play a significant role in the pathogenesis of ALS/PDC. Leveraging advanced induced pluripotent stem cell technology, our team cultivated multiple astrocyte lines to further investigate the Japanese variant of ALS/PDC (Kii ALS/PDC). CHCHD2 emerged as a significantly dysregulated gene when disease astrocytes were compared to healthy controls. Our analyses also revealed imbalances in the activation of specific pathways: those associated with astrocytic cilium dysfunction, known to be involved in neurodegeneration, and those related to major neurological disorders, including classical ALS and PD. Further in-depth examinations revealed abnormalities in the mitochondrial morphology and metabolic processes of the affected astrocytes. A particularly striking observation was the reduced expression of CHCHD2 in the spinal cord, motor cortex, and oculomotor nuclei of patients with Kii ALS/PDC. In summary, our findings suggest a potential reduction in the support Kii ALS/PDC astrocytes provide to neurons, emphasizing the need to explore the role of CHCHD2 in maintaining mitochondrial health and its implications for the disease.


Subject(s)
Amyotrophic Lateral Sclerosis , Astrocytes , DNA-Binding Proteins , Mitochondrial Proteins , Transcription Factors , Astrocytes/pathology , Astrocytes/metabolism , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Humans , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondria/pathology , Mitochondria/metabolism , Male , Female , Middle Aged , Aged
12.
Stem Cells ; 41(6): 603-616, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37029780

ABSTRACT

Despite developing neurosurgical procedures, few treatment options have achieved functional recovery from traumatic brain injury (TBI). Neural stem/progenitor cells (NS/PCs) may produce a long-term effect on neurological recovery. Although induced pluripotent stem cells (iPSCs) can overcome ethical and practical issues of human embryonic or fetal-derived tissues in clinical applications, the tumorigenicity of iPSC-derived populations remains an obstacle to their safe use in regenerative medicine. We herein established a novel treatment strategy for TBI using iPSCs expressing the enzyme-prodrug gene yeast cytosine deaminase-uracil phosphoribosyl transferase (yCD-UPRT). NS/PCs derived from human iPSCs displayed stable and high transgene expression of yCD-UPRT following CRISPR/Cas9-mediated genome editing. In vivo bioluminescent imaging and histopathological analysis demonstrated that NS/PCs concentrated around the damaged cortex of the TBI mouse model. During the subacute phase, performances in both beam walking test and accelerating rotarod test were significantly improved in the treatment group transplanted with genome-edited iPSC-derived NS/PCs compared with the control group. The injury area visualized by extravasation of Evans blue was smaller in the treatment group compared with the control group, suggesting the prevention of secondary brain injury. During the chronic phase, cerebral atrophy and ventricle enlargement were significantly less evident in the treatment group. Furthermore, after 5-fluorocytosine (5-FC) administration, 5-fluorouracil converted from 5-FC selectively eliminated undifferentiated NS/PCs while preserving the adjacent neuronal structures. NS/PCs expressing yCD-UPRT can be applied for safe regenerative medicine without the concern for tumorigenesis.


Subject(s)
Brain Injuries, Traumatic , Induced Pluripotent Stem Cells , Neural Stem Cells , Neuroprotective Agents , Mice , Animals , Humans , Induced Pluripotent Stem Cells/metabolism , Neuroprotective Agents/metabolism , Neural Stem Cells/metabolism , Neurons , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/therapy
13.
Brain ; 146(1): 13-19, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36004509

ABSTRACT

Using patient-derived induced pluripotent stem cells, neurodegenerative disease phenotypes have been recapitulated and their pathogenesis analysed leading to significant progress in drug screening. In amyotrophic lateral sclerosis, high-throughput screening using induced pluripotent stem cells-derived motor neurons has identified candidate drugs. Owing to induced pluripotent stem cell-based drug evaluation/screening, three compounds, retigabine, ropinirole and bosutinib, have progressed to clinical trials. Retigabine blocks hyperexcitability and improves survival in amyotrophic lateral sclerosis patient-derived motor neurons. In a randomized clinical trial (n = 65), treatment with retigabine reduced neuronal excitability after 8 weeks. Ropinirole, identified in a high-throughput screening, attenuates pathological phenotypes in patient-derived motor neurons. In a trial limited by a small sample size (n = 20), ropinirole was tolerable and had clinical benefits on function and survival. A phase 1 study of bosutinib has reported safety and tolerability for 12 weeks. Thus, these clinical trials show safety and positive effects and confirm the reliability of stem cell-based drug discovery. This novel strategy leads to reduced costs and time when compared to animal testing and opens new avenues for therapy in intractable diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , Induced Pluripotent Stem Cells , Neurodegenerative Diseases , Animals , Amyotrophic Lateral Sclerosis/genetics , Induced Pluripotent Stem Cells/pathology , Neurodegenerative Diseases/pathology , Drug Evaluation, Preclinical , Reproducibility of Results
14.
Cereb Cortex ; 33(9): 5148-5162, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36222604

ABSTRACT

Common marmosets are promising laboratory animals for the study of higher brain functions. Although there are many opportunities to use sedatives and anesthetics in resting brain function measurements in marmosets, their effects on the resting-state network remain unclear. In this study, the effects of sedatives or anesthetics such as midazolam, dexmedetomidine, co-administration of isoflurane and dexmedetomidine, propofol, alfaxalone, isoflurane, and sevoflurane on the resting brain function in common marmosets were evaluated using independent component analysis, dual regression analysis, and graph-theoretic analysis; and the sedatives or anesthetics suitable for the evaluation of resting brain function were investigated. The results show that network preservation tendency under light sedative with midazolam and dexmedetomidine is similar regardless of the type of target receptor. Moreover, alfaxalone, isoflurane, and sevoflurane have similar effects on resting state brain function, but only propofol exhibits different tendencies, as resting brain function is more preserved than it is following the administration of the other anesthetics. Co-administration of isoflurane and dexmedetomidine shows middle effect between sedatives and anesthetics.


Subject(s)
Anesthetics , Dexmedetomidine , Isoflurane , Propofol , Animals , Hypnotics and Sedatives/pharmacology , Callithrix , Isoflurane/pharmacology , Sevoflurane/pharmacology , Midazolam/pharmacology , Dexmedetomidine/pharmacology , Anesthetics/pharmacology , Brain
15.
Endocr J ; 71(2): 153-169, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38191197

ABSTRACT

Obesity and aging are major risk factors for several life-threatening diseases. Accumulating evidence from both rodents and humans suggests that the levels of nicotinamide adenine dinucleotide (NAD+), a regulator of many biological processes, declines in multiple organs and tissues with aging and obesity. Administration of an NAD+ intermediate, nicotinamide mononucleotide (NMN), replenishes intracellular NAD+ levels and mitigates aging- and obesity-associated derangements in animal models. In this human clinical study, we aimed to investigate the safety and effects of 8-week oral administration of NMN on biochemical, metabolic, ophthalmologic, and sleep quality parameters as well as on chronological alterations in NAD+ content in peripheral tissues. An 8-week, single-center, single-arm, open-label clinical trial was conducted. Eleven healthy, middle-aged Japanese men received two 125-mg NMN capsules once daily before breakfast. The 8-week NMN supplementation regimen was well-tolerated; NAD+ levels in peripheral blood mononuclear cells increased over the course of NMN administration. In participants with insulin oversecretion after oral glucose loading, NMN modestly attenuated postprandial hyperinsulinemia, a risk factor for coronary artery disease (n = 3). In conclusion, NMN overall safely and effectively boosted NAD+ biosynthesis in healthy, middle-aged Japanese men, showing its potential for alleviating postprandial hyperinsulinemia.


Subject(s)
Hyperinsulinism , NAD , Male , Middle Aged , Animals , Humans , NAD/metabolism , Nicotinamide Mononucleotide/metabolism , Leukocytes, Mononuclear/metabolism , Japan , Obesity , Sleep , Dietary Supplements
16.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33443207

ABSTRACT

The developing brain is under the risk of exposure to a multitude of environmental stressors. While perinatal exposure to excessive levels of environmental stress is responsible for a wide spectrum of neurological and psychiatric conditions, the developing brain is equipped with intrinsic cell protection, the mechanisms of which remain unknown. Here we show, using neonatal mouse as a model system, that primary cilia, hair-like protrusions from the neuronal cell body, play an essential role in protecting immature neurons from the negative impacts of exposure to environmental stress. More specifically, we found that primary cilia prevent the degeneration of dendritic arbors upon exposure to alcohol and ketamine, two major cell stressors, by activating cilia-localized insulin-like growth factor 1 receptor and downstream Akt signaling. We also found that activation of this pathway inhibits Caspase-3 activation and caspase-mediated cleavage/fragmentation of cytoskeletal proteins in stress-exposed neurons. These results indicate that primary cilia play an integral role in mitigating adverse impacts of environmental stressors such as drugs on perinatal brain development.


Subject(s)
Cilia/metabolism , Neural Stem Cells/metabolism , Prosencephalon/embryology , Animals , Animals, Newborn/metabolism , Brain/metabolism , Dendrites/metabolism , Female , Insulin-Like Growth Factor I/metabolism , Mice/embryology , Mice, Inbred C57BL , Neurons/metabolism , Pregnancy , Prenatal Exposure Delayed Effects , Prosencephalon/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
17.
J Neurosci ; 42(47): 8881-8896, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36261283

ABSTRACT

Fused in sarcoma (FUS) is a pathogenic RNA-binding protein in amyotrophic lateral sclerosis (ALS). We previously reported that FUS stabilizes Synaptic Ras-GTPase activating protein 1 (Syngap1) mRNA at its 3' untranslated region (UTR) and maintains spine maturation. To elucidate the pathologic roles of this mechanism in ALS patients, we identified the SYNGAP1 3'UTR variant rs149438267 in seven (four males and three females) out of 807 ALS patients at the FUS binding site from a multicenter cohort in Japan. Human-induced pluripotent stem cell (hiPSC)-derived motor neurons with the SYNGAP1 variant showed aberrant splicing, increased isoform α1 levels, and decreased isoform γ levels, which caused dendritic spine loss. Moreover, the SYNGAP1 variant excessively recruited FUS and heterogeneous nuclear ribonucleoprotein K (HNRNPK), and antisense oligonucleotides (ASOs) blocking HNRNPK altered aberrant splicing and ameliorated dendritic spine loss. These data suggest that excessive recruitment of RNA-binding proteins, especially HNRNPK, as well as changes in SYNGAP1 isoforms, are crucial for spine formation in motor neurons.SIGNIFICANCE STATEMENT It is not yet known which RNAs cause the pathogenesis of amyotrophic lateral sclerosis (ALS). We previously reported that Fused in sarcoma (FUS), a pathogenic RNA-binding protein in ALS, stabilizes synaptic Ras-GTPase activating protein 1 (Syngap1) mRNA at its 3' untranslated region (UTR) and maintains dendritic spine maturation. To elucidate whether this mechanism is crucial for ALS, we identified the SYNGAP1 3'UTR variant rs149438267 at the FUS binding site. Human-induced pluripotent stem cell (hiPSC)-derived motor neurons with the SYNGAP1 variant showed aberrant splicing, which caused dendritic spine loss along with excessive recruitment of FUS and heterogeneous nuclear ribonucleoprotein K (HNRNPK). Our findings that dendritic spine loss is because of excess recruitment of RNA-binding proteins provide a basis for the future exploration of ALS-related RNA-binding proteins.


Subject(s)
Amyotrophic Lateral Sclerosis , Sarcoma , Male , Female , Humans , Amyotrophic Lateral Sclerosis/metabolism , 3' Untranslated Regions/genetics , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Dendritic Spines/metabolism , Mutation , RNA-Binding Proteins/genetics , RNA, Messenger/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Sarcoma/genetics , ras GTPase-Activating Proteins/genetics
18.
Neuroimage ; 279: 120329, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37591477

ABSTRACT

Advancements in non-invasive brain analysis through novel approaches such as big data analytics and in silico simulation are essential for explaining brain function and associated pathologies. In this study, we extend the vector auto-regressive surrogate technique from a single multivariate time-series to group data using a novel Group Surrogate Data Generating Model (GSDGM). This methodology allowed us to generate biologically plausible human brain dynamics representative of a large human resting-state (rs-fMRI) dataset obtained from the Human Connectome Project. Simultaneously, we defined a novel similarity measure, termed the Multivariate Time-series Ensemble Similarity Score (MTESS). MTESS showed high accuracy and f-measure in subject identification, and it can directly compare the similarity between two multivariate time-series. We used MTESS to analyze both human and marmoset rs-fMRI data. Our results showed similarity differences between cortical and subcortical regions. We also conducted MTESS and state transition analysis between single and group surrogate techniques, and confirmed that a group surrogate approach can generate plausible group centroid multivariate time-series. Finally, we used GSDGM and MTESS for the fingerprint analysis of human rs-fMRI data, successfully distinguishing normal and outlier sessions. These new techniques will be useful for clinical applications and in silico simulation.


Subject(s)
Brain , Magnetic Resonance Imaging , Humans , Animals , Brain/diagnostic imaging , Callithrix , Computer Simulation , Time Factors
19.
Neuroimage ; 273: 120096, 2023 06.
Article in English | MEDLINE | ID: mdl-37031828

ABSTRACT

A comparison of neuroanatomical features of the brain between humans and our evolutionary relatives, nonhuman primates, is key to understanding the human brain system and the neural basis of mental and neurological disorders. Although most comparative MRI studies of human and nonhuman primate brains have been based on brains of primates that had been used as subjects in experiments, it is essential to investigate various species of nonhuman primates in order to elucidate and interpret the diversity of neuroanatomy features among humans and nonhuman primates. To develop a research platform for this purpose, it is necessary to harmonize the scientific contributions of studies with the standards of animal ethics, animal welfare, and the conservation of brain information for long-term continuation of the field. In previous research, we first developed a gated data-repository of anatomical images obtained using 9.4-T ex vivo MRI of postmortem brain samples from 12 nonhuman primate species, and which are stored at the Japan Monkey Centre. In the present study, as a second phase, we released a collection of T2-weighted images and diffusion tensor images obtained in nine species: white-throated capuchin, Bolivian squirrel monkey, stump-tailed macaque, Tibet monkey, Sykes' monkey, Assamese macaque, pig-tailed macaque, crested macaque, and chimpanzee. Our image repository should facilitate scientific discoveries in the field of comparative neuroscience. This repository can also promote animal ethics and animal welfare in experiments with nonhuman primate models by optimizing methods for in vivo and ex vivo MRI scanning of brains and supporting veterinary neuroradiological education. In addition, the repository is expected to contribute to conservation, preserving information about the brains of various primates, including endangered species, in a permanent digital form.


Subject(s)
Magnetic Resonance Imaging , Primates , Animals , Humans , Japan , Primates/anatomy & histology , Brain/diagnostic imaging , Brain/anatomy & histology , Macaca , Magnetic Resonance Spectroscopy , Neuroimaging
20.
J Neurochem ; 167(5): 603-614, 2023 12.
Article in English | MEDLINE | ID: mdl-37952981

ABSTRACT

It has been more than 10 years since the hopes for disease modeling and drug discovery using induced pluripotent stem cell (iPSC) technology boomed. Recently, clinical trials have been conducted with drugs identified using this technology, and some promising results have been reported. For amyotrophic lateral sclerosis (ALS), a devastating neurodegenerative disease, several groups have identified candidate drugs, ezogabine (retigabine), bosutinib, and ropinirole, using iPSCs-based drug discovery, and clinical trials using these drugs have been conducted, yielding interesting results. In our previous study, an iPSCs-based drug repurposing approach was utilized to show the potential of ropinirole hydrochloride (ROPI) in reducing ALS-specific pathological phenotypes. Recently, a phase 1/2a trial was conducted to investigate the effects of ropinirole on ALS further. This double-blind, randomized, placebo-controlled study confirmed the safety and tolerability of and provided evidence of its ability to delay disease progression and prolong the time to respiratory failure in ALS patients. Furthermore, in the reverse translational research, in vitro characterization of patient-derived iPSCs-motor neurons (MNs) mimicked the therapeutic effects of ROPI in vivo, suggesting the potential application of this technology to the precision medicine of ALS. Interestingly, RNA-seq data showed that ROPI treatment suppressed the sterol regulatory element-binding protein 2-dependent cholesterol biosynthesis pathway. Therefore, this pathway may be involved in the therapeutic effect of ROPI on ALS. The possibility that this pathway may be involved in the therapeutic effect of ALS was demonstrated. Finally, new future strategies for ALS using iPSCs technology will be discussed in this paper.


Subject(s)
Amyotrophic Lateral Sclerosis , Induced Pluripotent Stem Cells , Neurodegenerative Diseases , Humans , Amyotrophic Lateral Sclerosis/metabolism , Induced Pluripotent Stem Cells/metabolism , Drug Evaluation, Preclinical , Neurodegenerative Diseases/metabolism , Translational Research, Biomedical , Randomized Controlled Trials as Topic
SELECTION OF CITATIONS
SEARCH DETAIL