12 results on '"Taito Matsuda"'
Search Results
2. Np95/Uhrf1 regulates tumor suppressor gene expression of neural stem/precursor cells, contributing to neurogenesis in the adult mouse brain
- Author
-
Naoya Murao, Masahiro Mutoh, Hirofumi Noguchi, Taito Matsuda, Haruhiko Koseki, Masakazu Namihira, Shuzo Matsubara, Kinichi Nakashima, and Tetsuji Mutoh
- Subjects
0301 basic medicine ,Tumor suppressor gene ,Neurogenesis ,Ubiquitin-Protein Ligases ,Subventricular zone ,Mice, Transgenic ,Biology ,Hippocampal formation ,Hippocampus ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Neural Stem Cells ,Precursor cell ,medicine ,Animals ,Genes, Tumor Suppressor ,Nuclear protein ,Cells, Cultured ,Cyclin-Dependent Kinase Inhibitor p16 ,Cell Proliferation ,General Neuroscience ,Dentate gyrus ,Nuclear Proteins ,Cell Differentiation ,General Medicine ,humanities ,Cell biology ,Adult Stem Cells ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Dentate Gyrus ,DNA methylation ,CCAAT-Enhancer-Binding Proteins ,Tumor Suppressor Protein p53 ,030217 neurology & neurosurgery - Abstract
Adult neurogenesis is a process of generating new neurons from neural stem/precursor cells (NS/PCs) in restricted adult brain regions throughout life. It is now generally known that adult neurogenesis in the hippocampal dentate gyrus (DG) and subventricular zone participates in various higher brain functions, such as learning and memory formation, olfactory discrimination and repair after brain injury. However, the mechanisms underlying adult neurogenesis remain to be fully understood. Here, we show that Nuclear protein 95 KDa (Np95, also known as UHRF1 or ICBP90), which is an essential protein for maintaining DNA methylation during cell division, is involved in multiple processes of adult neurogenesis. Specific ablation of Np95 in adult NS/PCs (aNS/PCs) led to a decrease in their proliferation and an impairment of neuronal differentiation and to suppression of neuronal maturation associated with the impairment of dendritic formation in the hippocampal DG. We also found that deficiency of Np95 in NS/PCs increased the expression of tumor suppressor genes p16 and p53, and confirmed that expression of these genes in NS/PCs recapitulates the phenotype of Np95-deficient NS/PCs. Taken together, our findings suggest that Np95 plays an essential role in proliferation and differentiation of aNS/PCs through the regulation of tumor suppressor gene expression in adult neurogenesis.
- Published
- 2019
- Full Text
- View/download PDF
3. Regulation of Adult Mammalian Neural Stem Cells and Neurogenesis by Cell Extrinsic and Intrinsic Factors
- Author
-
Kinichi Nakashima, Shuzo Matsubara, and Taito Matsuda
- Subjects
QH301-705.5 ,injury ,Neurogenesis ,neurons ,microglia ,Endogeny ,Review ,Biology ,Regenerative Medicine ,Regenerative medicine ,Hippocampus ,Mice ,Neural Stem Cells ,medicine ,Animals ,Humans ,neurodegenerative diseases ,Cell Lineage ,Biology (General) ,Stem Cell Niche ,Tissue homeostasis ,Cell Proliferation ,Microglia ,astrocytes ,Brain ,Cell Differentiation ,General Medicine ,direct reprogramming ,Neural stem cell ,Extracellular Matrix ,Adult Stem Cells ,medicine.anatomical_structure ,nervous system ,Stem cell ,Nerve Net ,Transcriptome ,Neuroscience ,Reprogramming ,Signal Transduction - Abstract
Tissue-specific stem cells give rise to new functional cells to maintain tissue homeostasis and restore damaged tissue after injury. To ensure proper brain functions in the adult brain, neural stem cells (NSCs) continuously generate newborn neurons that integrate into pre-existing neuronal networks. Proliferation, as well as neurogenesis of NSCs, are exquisitely controlled by extrinsic and intrinsic factors, and their underlying mechanisms have been extensively studied with the goal of enhancing the neurogenic capacity of NSCs for regenerative medicine. However, neurogenesis of endogenous NSCs alone is insufficient to completely repair brains damaged by neurodegenerative diseases and/or injury because neurogenic areas are limited and few neurons are produced in the adult brain. An innovative approach towards replacing damaged neurons is to induce conversion of non-neuronal cells residing in injured sites into neurons by a process referred to as direct reprogramming. This review describes extrinsic and intrinsic factors controlling NSCs and neurogenesis in the adult brain and discusses prospects for their applications. It also describes direct neuronal reprogramming technology holding promise for future clinical applications.
- Published
- 2021
4. Prior Treatment with Anti-High Mobility Group Box-1 Antibody Boosts Human Neural Stem Cell Transplantation-Mediated Functional Recovery After Spinal Cord Injury
- Author
-
Kinichi Nakashima, Naohiro Uezono, Taito Matsuda, Yusuke Fujimoto, Yicheng Zhu, Shuji Mori, Tetsuro Yasui, Takao Setoguchi, Masahiro Nishibori, Masahide Nakajo, Hideo Takahashi, Masahiko Abematsu, and Setsuro Komiya
- Subjects
0301 basic medicine ,Cord ,Neurite ,Mice, SCID ,Biology ,HMGB1 ,03 medical and health sciences ,Neural Stem Cells ,Mice, Inbred NOD ,medicine ,Biological neural network ,Animals ,Humans ,HMGB1 Protein ,Spinal cord injury ,Cells, Cultured ,Spinal Cord Injuries ,Cell Differentiation ,Recovery of Function ,Cell Biology ,medicine.disease ,Neural stem cell ,Cell biology ,Transplantation ,Disease Models, Animal ,030104 developmental biology ,nervous system ,biology.protein ,Molecular Medicine ,Stem cell ,Stem Cell Transplantation ,Developmental Biology - Abstract
Together with residual host neurons, transplanted neural stem cell (NSC)-derived neurons play a critical role in reconstructing disrupted neural circuits after spinal cord injury (SCI). Since a large number of tracts are disrupted and the majority of host neurons die around the lesion site as the damage spreads, minimizing this spreading and preserving the lesion site are important for attaining further improvements in reconstruction. High mobility group box-1 (HMGB1) is a damage-associated molecular pattern protein that triggers sterile inflammation after tissue injury. In the ischemic and injured brain, neutralization of HMGB1 with a specific antibody reportedly stabilizes the blood-brain barrier, suppresses inflammatory cytokine expression, and improves functional recovery. Using a SCI model mouse, we here developed a combinatorial treatment for SCI: administering anti-HMGB1 antibody prior to transplantation of NSCs derived from human induced pluripotent stem cells (hiPSC-NSCs) yielded a dramatic improvement in locomotion recovery after SCI. Even anti-HMGB1 antibody treatment alone alleviated blood-spinal cord barrier disruption and edema formation, and increased the number of neurites from spared axons and the survival of host neurons, resulting in functional recovery. However, this recovery was greatly enhanced by the subsequent hiPSC-NSC transplantation, reaching an extent that has never before been reported. We also found that this improved recovery was directly associated with connections established between surviving host neurons and transplant-derived neurons. Taken together, our results highlight combinatorial treatment with anti-HMGB1 antibody and hiPSC-NSC transplantation as a promising novel therapy for SCI.
- Published
- 2018
- Full Text
- View/download PDF
5. NEUROD1 Instructs Neuronal Conversion in Non-Reactive Astrocytes
- Author
-
Brian K. Kaspar, Mauro Giacca, Taito Matsuda, Kinichi Nakashima, Carlos Henrique Miranda, Jenny Hsieh, Rebecca Brulet, and Ling Zhang
- Subjects
0301 basic medicine ,Male ,transdifferentiation ,Cellular differentiation ,striatum ,neurons ,Striatum ,Biochemistry ,Mice ,0302 clinical medicine ,Basic Helix-Loop-Helix Transcription Factors ,lcsh:QH301-705.5 ,Cells, Cultured ,NeuroD ,lcsh:R5-920 ,Transdifferentiation ,Cell Differentiation ,Dependovirus ,Cellular Reprogramming ,3. Good health ,Cell biology ,medicine.anatomical_structure ,cortex ,Blood-Brain Barrier ,Cell Transdifferentiation ,AAV9 ,lcsh:Medicine (General) ,Genetic Vectors ,Mice, Transgenic ,Nerve Tissue Proteins ,Biology ,Blood–brain barrier ,03 medical and health sciences ,Report ,Genetics ,medicine ,Animals ,Transcription factor ,astrocytes ,reprogramming ,Cell Biology ,Corpus Striatum ,Rats ,030104 developmental biology ,nervous system ,lcsh:Biology (General) ,NEUROD1 ,Immunology ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Summary Currently, all methods for converting non-neuronal cells into neurons involve injury to the brain; however, whether neuronal transdifferentiation can occur long after the period of insult remains largely unknown. Here, we use the transcription factor NEUROD1, previously shown to convert reactive glial cells to neurons in the cortex, to determine whether astrocyte-to-neuron transdifferentiation can occur under physiological conditions. We utilized adeno-associated virus 9 (AAV9), which crosses the blood-brain barrier without injury, to deliver NEUROD1 to astrocytes through an intravascular route. Interestingly, we found that a small, but significant number of non-reactive astrocytes converted to neurons in the striatum, but not the cortex. Moreover, astrocytes cultured to minimize their proliferative potential also exhibited limited neuronal transdifferentiation with NEUROD1 expression. Our results show that a single transcription factor can induce astrocyte-to-neuron conversion under physiological conditions, potentially facilitating future clinical approaches long after the acute injury phase., Highlights • Intravascular AAV9 efficiently targets astrocytes in the cortex and striatum • NEUROD1 can convert a small, but significant number of astrocytes to neurons in vivo • NEUROD1 induces limited neuronal transdifferentiation in vitro • Injury-related factors may be important in neuronal conversion strategies, In this article, Hsieh and colleagues use AAV9 viral infection to selectively target and overexpress NEUROD1 in astrocytes in the absence of injury to the CNS. The authors show that NEUROD1 is capable of converting a small, but significant number of astrocytes to neurons in vivo in the absence of reactive gliosis.
- Published
- 2017
6. DNA Methyltransferase 1 Is Indispensable for Development of the Hippocampal Dentate Gyrus
- Author
-
Masakazu Namihira, Ayaka Kimura, Taito Matsuda, Kinichi Nakashima, Hirofumi Noguchi, and Naoya Murao
- Subjects
DNA (Cytosine-5-)-Methyltransferase 1 ,Doublecortin Domain Proteins ,0301 basic medicine ,Neurogenesis ,Green Fluorescent Proteins ,Mice, Transgenic ,Nerve Tissue Proteins ,Hippocampal formation ,environment and public health ,Subgranular zone ,Nestin ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Neural Stem Cells ,medicine ,Animals ,DNA (Cytosine-5-)-Methyltransferases ,Reelin ,Cells, Cultured ,reproductive and urinary physiology ,Neurons ,Mice, Inbred ICR ,biology ,General Neuroscience ,Dentate gyrus ,Neuropeptides ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Articles ,Embryo, Mammalian ,Granule cell ,Embryonic stem cell ,Neural stem cell ,Reelin Protein ,030104 developmental biology ,medicine.anatomical_structure ,Animals, Newborn ,nervous system ,Dentate Gyrus ,embryonic structures ,biology.protein ,Microtubule-Associated Proteins ,Neuroscience ,030217 neurology & neurosurgery ,Intracellular - Abstract
Development of the hippocampal dentate gyrus (DG) in the mammalian brain is achieved through multiple processes during late embryonic and postnatal stages, with each developmental step being strictly governed by extracellular cues and intracellular mechanisms. Here, we show that the maintenance DNA methyltransferase 1 (Dnmt1) is critical for development of the DG in the mouse. Deletion ofDnmt1in neural stem cells (NSCs) at the beginning of DG development led to a smaller size of the granule cell layer in the DG. NSCs lackingDnmt1failed to establish proper radial processes or to migrate into the subgranular zone, resulting in aberrant neuronal production in the molecular layer of the DG and a reduction of integrated neurons in the granule cell layer. Interestingly, prenatal deletion ofDnmt1in NSCs affected not only the developmental progression of the DG but also the properties of NSCs maintained into adulthood:Dnmt1-deficient NSCs displayed impaired neurogenic ability and proliferation. We also found thatDnmt1deficiency in NSCs decreased the expression of Reelin signaling components in the developing DG and increased that of the cell cycle inhibitorsp21andp57in the adult DG. Together, these findings led us to propose that Dnmt1 functions as a key regulator to ensure the proper development of the DG, as well as the proper status of NSCs maintained into adulthood, by modulating extracellular signaling and intracellular mechanisms.SIGNIFICANCE STATEMENTHere, we provide evidence that Dnmt1 is required for the proper development of the hippocampal dentate gyrus (DG). Deletion ofDnmt1in neural stem cells (NSCs) at an early stage of DG development impaired the ability of NSCs to establish secondary radial glial scaffolds and to migrate into the subgranular zone of the DG, leading to aberrant neuronal production in the molecular layer, increased cell death, and decreased granule neuron production. Prenatal deletion ofDnmt1in NSCs also induced defects in the proliferation and neurogenic ability of adult NSCs. Furthermore, we found that Dnmt1 regulates the expression of key extracellular signaling components during developmental stages while modulating intracellular mechanisms for proliferation and neuronal production of NSCs in the adult.
- Published
- 2016
- Full Text
- View/download PDF
7. Ectopic neurogenesis induced by prenatal antiepileptic drug exposure augments seizure susceptibility in adult mice
- Author
-
Kiyoko Kato, Yukiko Nagaishi, Atsuhiko Sakai, Taito Matsuda, Hiroyoshi Doi, and Kinichi Nakashima
- Subjects
0301 basic medicine ,Hippocampus ,Epilepsy ,neural stem cell ,Cxcr4 ,Mice ,Random Allocation ,0302 clinical medicine ,Neural Stem Cells ,Cell Movement ,Pregnancy ,Medicine ,Cells, Cultured ,ectopic neurogenesis ,Neurons ,Valproic Acid ,Multidisciplinary ,Neurogenesis ,Gene Expression Regulation, Developmental ,Biological Sciences ,Neural stem cell ,Current Literature in Basic Science ,In utero ,Prenatal Exposure Delayed Effects ,lipids (amino acids, peptides, and proteins) ,Anticonvulsants ,Female ,Disease Susceptibility ,medicine.drug ,medicine.medical_specialty ,Receptors, CXCR4 ,Offspring ,Physical Exertion ,Gestational Age ,Nerve Tissue Proteins ,03 medical and health sciences ,Seizures ,Internal medicine ,Animals ,RNA, Messenger ,Progenitor cell ,business.industry ,medicine.disease ,Rats ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Dentate Gyrus ,epilepsy ,business ,Transcriptome ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Significance Recent clinical studies suggest that environmental insults, such as valproic acid (VPA) exposure, in utero can have adverse effects on brain function of the offspring in later life, although the underlying mechanisms of these impairments remain poorly understood. By focusing on the property of neural stem/progenitor cells (NS/PCs) residing in the adult hippocampus, we identified the mechanism of increased seizure sensitivity in prenatally VPA-exposed adult mice. Furthermore, we found that voluntary exercise can overcome the adverse effects through normalizing VPA-induced transcriptome alterations in NS/PCs. We believe that our study provides insights for further understanding and developing treatment strategies for neurological disorders induced by prenatal environmental insults., Epilepsy is a neurological disorder often associated with seizure that affects ∼0.7% of pregnant women. During pregnancy, most epileptic patients are prescribed antiepileptic drugs (AEDs) such as valproic acid (VPA) to control seizure activity. Here, we show that prenatal exposure to VPA in mice increases seizure susceptibility in adult offspring through mislocalization of newborn neurons in the hippocampus. We confirmed that neurons newly generated from neural stem/progenitor cells (NS/PCs) are integrated into the granular cell layer in the adult hippocampus; however, prenatal VPA treatment altered the expression in NS/PCs of genes associated with cell migration, including CXC motif chemokine receptor 4 (Cxcr4), consequently increasing the ectopic localization of newborn neurons in the hilus. We also found that voluntary exercise in a running wheel suppressed this ectopic neurogenesis and countered the enhanced seizure susceptibility caused by prenatal VPA exposure, probably by normalizing the VPA-disrupted expression of multiple genes including Cxcr4 in adult NS/PCs. Replenishing Cxcr4 expression alone in NS/PCs was sufficient to overcome the aberrant migration of newborn neurons and increased seizure susceptibility in VPA-exposed mice. Thus, prenatal exposure to an AED, VPA, has a long-term effect on the behavior of NS/PCs in offspring, but this effect can be counteracted by a simple physical activity. Our findings offer a step to developing strategies for managing detrimental effects in offspring exposed to VPA in utero.
- Published
- 2018
8. Expression of DNMT1 in neural stem/precursor cells is critical for survival of newly generated neurons in the adult hippocampus
- Author
-
Hirofumi Noguchi, Masakazu Namihira, Taito Matsuda, Ayaka Kimura, Kinichi Nakashima, and Naoya Murao
- Subjects
DNA (Cytosine-5-)-Methyltransferase 1 ,Cell Survival ,Neurogenesis ,Cellular differentiation ,Hippocampus ,Mice, Transgenic ,Biology ,Mice ,Neural Stem Cells ,Precursor cell ,Animals ,DNA (Cytosine-5-)-Methyltransferases ,Cell Proliferation ,Neurons ,General Neuroscience ,Dentate gyrus ,Cell Differentiation ,General Medicine ,Neural stem cell ,Adult Stem Cells ,nervous system ,Dentate Gyrus ,DNA methylation ,Neuroscience ,Adult stem cell - Abstract
Adult neurogenesis persists throughout life in the dentate gyrus (DG) of the hippocampus, and its importance has been highlighted in hippocampus-dependent learning and memory. Adult neurogenesis consists of multiple processes: maintenance and neuronal differentiation of neural stem/precursor cells (NS/PCs), followed by survival and maturation of newborn neurons and their integration into existing neuronal circuitry. However, the mechanisms that govern these processes remain largely unclear. Here we show that DNA methyltransferase 1 (DNMT1), an enzyme responsible for the maintenance of DNA methylation, is highly expressed in proliferative cells in the adult DG and plays an important role in the survival of newly generated neurons. Deletion of Dnmt1 in adult NS/PCs (aNS/PCs) did not affect the proliferation and differentiation of aNS/PCs per se. However, it resulted in a decrease of newly generated mature neurons, probably due to gradual cell death after aNS/PCs differentiated into neurons in the hippocampus. Interestingly, loss of DNMT1 in post-mitotic neurons did not influence their survival. Taken together, these findings suggest that the presence of DNMT1 in aNS/PCs is crucial for the survival of newly generated neurons, but is dispensable once they accomplish neuronal differentiation in the adult hippocampus.
- Published
- 2015
- Full Text
- View/download PDF
9. HMGB2 expression is associated with transition from a quiescent to an activated state of adult neural stem cells
- Author
-
Ayaka, Kimura, Taito, Matsuda, Atsuhiko, Sakai, Naoya, Murao, and Kinichi, Nakashima
- Subjects
Neurons ,Adult Stem Cells ,Mice ,Neural Stem Cells ,Lateral Ventricles ,Neurogenesis ,Animals ,HMGB2 Protein ,Hippocampus - Abstract
Although quiescent neural stem cells (NSCs) in the adult hippocampus proliferate in response to neurogenic stimuli and subsequently give rise to new neurons continuously throughout life, misregulation of NSCs in pathological conditions, including aging, leads to the impairment of learning and memory. High mobility group B family 1 (HMGB1) and HMGB2, HMG family proteins that function as transcriptional activators through the modulation of chromatin structure, have been assumed to play some role in the regulation of adult NSCs; however, their precise functions and even expression patterns in the adult hippocampus remain elusive.Here we show that expression of HMGB2 but not HMGB1 is restricted to the subset of NSCs and their progenitors. Furthermore, running, a well-known positive neurogenic stimulus, increased the proliferation of HMGB2-expressing cells, whereas aging was accompanied by a marked decrease in these cells. Intriguingly, HMGB2-expressing quiescent NSCs, which were shifted toward the proliferative state, were decreased as aging progressed.HMGB2 expression is strongly associated with transition from the quiescent to the proliferative state of NSCs, supporting the possibility that HMGB2 is involved in the regulation of adult neurogenesis and can be used as a novel marker to identify NSCs primed for activation in the adult hippocampus. Developmental Dynamics 247:229-238, 2018. © 2017 Wiley Periodicals, Inc.
- Published
- 2017
10. Epigenetic regulation of neural stem cell fate during corticogenesis
- Author
-
Berry Juliandi, Chai MuhChyi, Taito Matsuda, and Kinichi Nakashima
- Subjects
Cerebral Cortex ,Epigenomics ,Neurons ,Multipotent Stem Cells ,Neurogenesis ,Neural tube ,EMX1 ,Biology ,Neural stem cell ,nervous system diseases ,Corticogenesis ,medicine.anatomical_structure ,nervous system ,Developmental Neuroscience ,Cerebral cortex ,medicine ,Animals ,biological phenomena, cell phenomena, and immunity ,Neuroscience ,Neural cell ,reproductive and urinary physiology ,Developmental Biology ,Gliogenesis - Abstract
The cerebral cortex comprises over three quarters of the brain, and serves as structural basis for the sophisticated perceptual and cognitive functions. It develops from common multipotent neural stem cells (NSCs) that line the neural tube. Development of the NSCs encompasses sequential phases of progenitor expansion, neurogenesis, and gliogenesis along with the progression of developmental stages. Interestingly, NSCs steadfastly march through all of these phases and give rise to specific neural cell types in a temporally defined and highly predictable manner. Herein, we delineate the intrinsic and extrinsic factors that dictate the progression and tempo of NSC differentiation during cerebral cortex development, and how epigenetic modifications contribute to the dynamic properties of NSCs.
- Published
- 2013
- Full Text
- View/download PDF
11. TLR9 signalling in microglia attenuates seizure-induced aberrant neurogenesis in the adult hippocampus
- Author
-
Shizuo Akira, Taito Matsuda, Yuki Katano, Naoya Murao, Kinichi Nakashima, Taro Kawai, Berry Juliandi, and Jun Kohyama
- Subjects
Male ,hippocampus ,Neurogenesis ,seizure ,General Physics and Astronomy ,Hippocampus ,Biology ,Severity of Illness Index ,Article ,General Biochemistry, Genetics and Molecular Biology ,Epilepsy ,Mice ,Cognition ,Seizures ,Toll-like receptor ,medicine ,Animals ,Cognitive Dysfunction ,Cognitive decline ,Progenitor ,Mice, Knockout ,Neurons ,Multidisciplinary ,Innate immune system ,Membrane Glycoproteins ,Microglia ,Tumor Necrosis Factor-alpha ,General Chemistry ,medicine.disease ,Immunity, Innate ,Mice, Inbred C57BL ,medicine.anatomical_structure ,nervous system ,Gene Expression Regulation ,Toll-Like Receptor 7 ,Toll-Like Receptor 9 ,Immunology ,Commentary ,epilepsy ,Tumor necrosis factor alpha ,neural stem/progenitor cell ,Signal Transduction - Abstract
Pathological conditions such as epilepsy cause misregulation of adult neural stem/progenitor populations in the adult hippocampus in mice, and the resulting abnormal neurogenesis leads to impairment in learning and memory. However, how animals cope with abnormal neurogenesis remains unknown. Here we show that microglia in the mouse hippocampus attenuate convulsive seizure-mediated aberrant neurogenesis through the activation of Toll-like receptor 9 (TLR9), an innate immune sensor known to recognize microbial DNA and trigger inflammatory responses. We found that microglia sense self-DNA from degenerating neurons following seizure, and secrete tumour necrosis factor-α, resulting in attenuation of aberrant neurogenesis. Furthermore, TLR9 deficiency exacerbated seizure-induced cognitive decline and recurrent seizure severity. Our findings thus suggest the existence of bidirectional communication between the innate immune and nervous systems for the maintenance of adult brain integrity., Epileptic seizures generate aberrant neurogenesis in the adult mouse hippocampal region but how animals cope with abnormal neurogenesis remains unknown. Here the authors show that microglia are activated through TLR9 signaling and that this leads to sustained expression of TNF-α which attenuates induced aberrant neurogenesis.
- Published
- 2014
12. Functional regulation of transient receptor potential canonical 7 by cGMP-dependent protein kinase Iα
- Author
-
Keizo Yuasa, Akihiko Tsuji, and Taito Matsuda
- Subjects
Recombinant Fusion Proteins ,Molecular Sequence Data ,Biology ,Mitogen-activated protein kinase kinase ,MAP2K7 ,TRPC3 ,Chlorocebus aethiops ,Cyclic GMP-Dependent Protein Kinases ,Animals ,Humans ,Amino Acid Sequence ,Calcium Signaling ,Phosphorylation ,Protein kinase A ,Cyclic AMP Response Element-Binding Protein ,Cyclic GMP-Dependent Protein Kinase Type I ,Enzyme Assays ,TRPC Cation Channels ,Kinase ,Cell Biology ,Molecular biology ,Isoenzymes ,HEK293 Cells ,COS Cells ,Mutagenesis, Site-Directed ,Carbachol ,Signal transduction ,cGMP-dependent protein kinase ,Protein Binding - Abstract
The cGMP/cGMP-dependent protein kinase (cGK) signaling pathway is implicated in the functional regulation of intracellular calcium levels. In the present study, we investigated the regulation of transient receptor potential canonical 7 (TRPC7) by the cGMP/cGK-I pathway. TRPC7 contains three putative cGK phosphorylation sites (Arg-Arg/Lys-Xaa-Ser/Thr). However, the role of cGK-I in the regulation of TRPC7 activity remains unclear. In vitro and in vivo kinase assays have revealed that cGK-Iα phosphorylates mouse TRPC7 but not mouse TRPC3. Site-directed mutagenesis analysis revealed that TRPC7 was phosphorylated by cGK-Iα at threonine 15. Phosphorylation of TRPC7 significantly suppressed carbachol-induced calcium influx and CREB phosphorylation. Furthermore, co-immunoprecipitation assay demonstrated that cGK-Iα interacted with the ankyrin repeat domain in the N terminus of TRPC7. cGK-Iβ also bound to TRPC7, while the type II regulatory subunit of cAMP-dependent protein kinase did not bind. These data indicate that cGK-Iα interacts with and phosphorylates TRPC7, contributing to the quick and accurate regulation of calcium influx and CREB phosphorylation.
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.