Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kazuaki Yoshikawa is active.

Publication


Featured researches published by Kazuaki Yoshikawa.


Journal of Biological Chemistry | 1998

Necdin, A Postmitotic Neuron-specific Growth Suppressor, Interacts with Viral Transforming Proteins and Cellular Transcription Factor E2F1

Hideo Taniura; Naoko Taniguchi; Mizuki Hara; Kazuaki Yoshikawa

Necdin is a nuclear protein expressed in virtually all postmitotic neurons, and ectopic expression of this protein strongly suppresses the proliferation of NIH3T3 cells. Simian virus 40 large T antigen targets both p53 and the retinoblastoma protein (Rb) for cellular transformation. By analogy with the interactions of the large T antigen with these nuclear growth suppressors, we examined the ability of necdin to bind to the large T antigen. Necdin was co-immunoprecipitated with the large T antigen from the nuclear extract of necdin cDNA-transfected COS-1 cells. Yeast two-hybrid and in vitro binding analyses revealed that necdin bound to an amino-terminal region of the large T antigen, which encompasses the Rb-binding domain. Moreover, necdin bound to adenovirus E1A, another viral oncoprotein that forms a specific complex with Rb. We then examined the ability of necdin to bind to the transcription factor E2F1, a cellular Rb-binding factor involved in cell-cycle progression. Intriguingly, necdin, like Rb, bound to a carboxyl-terminal domain of E2F1, and repressed E2F-dependent transactivation in vivo. In addition, necdin suppressed the colony formation of Rb-deficient SAOS-2 osteosarcoma cells. These results suggest that necdin is a postmitotic neuron-specific growth suppressor that is functionally similar to Rb.


Neuroscience Research | 2000

Cell cycle regulators in neural stem cells and postmitotic neurons.

Kazuaki Yoshikawa

In the mammalian central nervous system, neurons withdraw from the cell cycle immediately after their differentiation from proliferative neuroepithelial cells. Even while postmitotic neurons remain in permanent mitotic quiescence, they express a number of cell cycle regulators required for cell cycle progression. This review focuses on the expression and functions of members of the retinoblastoma protein (Rb) family (Rb, p107, p130) and necdin, all of which are growth suppressors that interact with the viral oncoproteins and the E2F family proteins. These molecules are differentially expressed in proliferative neural progenitors and postmitotic neurons in the developing neuroepithelium in vivo and differentiating embryonal carcinoma cells in vitro. During neurogenesis, dysfunction of the Rb family proteins causes impaired neuronal differentiation accompanied by cell death (apoptosis). Thus, the Rb family proteins are essential for both terminal mitosis of neuronal progenitors and survival of nascent neurons. However, the Rb family proteins seem to be dispensable for the maintenance of the postmitotic state of terminally differentiated neurons. Necdin is expressed exclusively in postmitotic cells and may contribute to their permanent mitotic arrest. These cell cycle regulators coordinately act in the generation, survival and demise of postmitotic neurons.


Biochemical and Biophysical Research Communications | 1991

A novel brain-specific mRNA encoding nuclear protein (necdin) expressed in neurally differentiated embryonal carcinoma cells

Kei Maruyama; Mihoko Usami; Takako Aizawa; Kazuaki Yoshikawa

A novel DNA sequence has been isolated from a subtraction cDNA library of P19 embryonal carcinoma cells treated with retinoic acid which induces neural differentiation of the stem cells. The cDNA insert (4B) hybridized with a single 1.7 kb mRNA, whose abundance was markedly increased in P19 cells after retinoic acid treatment. The 1.7 kb mRNA was also expressed in the brain, but not in other non-neuronal tissues. A 1.6 kb cDNA insert (4BFL), which was cloned by screening another cDNA library with the 4B probe, encodes a novel protein sequence of 325 amino acids (Mr 36,831). The protein expressed in 4BFL-transfected COS cells was translocated into the nuclei as detected with antibodies against subsequences of the predicted protein. The antibodies stained the nuclei of neurally differentiated P19 cells but not of the undifferentiated stem cells. This novel mRNA encoding the nuclear protein, termed necdin, may represent a useful marker for the differentiation and development of brain cells.


Journal of Biological Chemistry | 1999

Physical and Functional Interactions of Neuronal Growth Suppressor Necdin with p53

Hideo Taniura; Kuniharu Matsumoto; Kazuaki Yoshikawa

Necdin is expressed in virtually all postmitotic neurons, and ectopic expression of this protein suppresses cell proliferation. Necdin, like the retinoblastoma protein, interacts with cell cycle promoting proteins such as simian virus 40 large T antigen, adenovirus E1A, and the transcription factor E2F1. Here we demonstrate that necdin interacts with the tumor suppressor protein p53 as well. The yeast two-hybrid and in vitro binding analyses revealed that necdin bound to a narrow region (amino acids 35–62) located between the MDM2-binding site and the proline-rich region in the amino-terminal domain of p53. The electrophoretic mobility shift assay showed that necdin supershifted a complex between p53 and its binding DNA, implying that the p53-necdin complex is competent for DNA binding. In p53-deficient osteosarcoma SAOS-2 cells, necdin markedly suppressed p53-dependent activation of the p21/WAF promoter. Necdin and p53 inhibited cell growth in an additive manner as assessed by the colony formation of SAOS-2 cells, suggesting that necdin does not affect p53-mediated growth suppression. On the other hand, necdin inhibited p53-induced apoptosis of osteosarcoma U2OS cells. Thus, necdin can be a growth suppressor that targets p53 and modulates its biological functions in postmitotic neurons.


Journal of Biological Chemistry | 1996

Structure and Expression of the Mouse Necdin Gene IDENTIFICATION OF A POSTMITOTIC NEURON-RESTRICTIVE CORE PROMOTER

Taichi Uetsuki; Keiich Takagi; Hiroko Sugiura; Kazuaki Yoshikawa

Necdin is a 325 amino acid residue protein encoded by a cDNA clone isolated from neurally differentiated embryonal carcinoma cells. In situ hybridization histochemistry revealed that necdin mRNA-containing cells in vivo coincided with postmitotic neurons in the mouse brain from early periods of neurogenesis until adulthood. To study the regulation of necdin gene expression, we have isolated and characterized the necdin gene from a mouse genomic DNA library. The necdin gene contains no intron, and its upstream region lacks canonical TATA and CAAT boxes. To assess promoter activity, the 5′-flanking sequence (844 base pairs) of the necdin gene was fused to the LacZ reporter gene and transiently transfected into retinoic acid-treated P19 embryonal carcinoma cells. Most of the transfectants expressing high levels of LacZ immunoreactivity were postmitotic neurons differentiated from P19 cells. Deletion analysis using luciferase reporter genes demonstrated that a neuron-restrictive core promoter is localized to positions −80 to −35, in which a G+C-rich domain and a putative binding site for transcription factors with PAS (per, arnt, and single-minded) dimerization domain are comprised. These results suggest that postmitotic neuron-restrictive expression of the necdin gene is mediated by the specific cis-acting elements and that this promoter is applicable to postmitotic neuron-targeted expression of various transgenic systems.


Biochemical and Biophysical Research Communications | 1991

Alzheimer's disease amyloid β-clipping enzyme (APP secretase): Identification, purification, and characterization of the enzyme

Kazuhiko Tagawa; Tatsuhide Kunishita; Kei Maruyama; Kazuaki Yoshikawa; Eiki Kominami; Takahide Tsuchiya; Koichi Suzuki; Takeshi Tabira; Hideo Sugita; Shoichi Ishiura

Alzheimers disease (AD) is the most frequent cause of dementia, although no genetic abnormality has been identified. Recent studies have elucidated the molecular defect in AD, including the abnormal deposition of amyloid beta peptide (beta/A4) in senile plaques of affected individuals. Normal brain contains the enzyme, APP secretase, which cleaves inside the beta/A4 portion of the precursor protein (APP); abnormal processing of APP occurs in AD brain. Until now, no evidence has been provided that APP secretase is an intracellular proteinase. We have now prepared two synthetic substrates of APP secretase, both of which contain the cleavage point and are much more sensitive than substrates previously available to identify APP secretase. Using these substrates, we found an intracellular proteinase that has APP secretase activity. This proteinase has been identified as cathepsin B.


Journal of Biological Chemistry | 2000

The Postmitotic Growth Suppressor Necdin Interacts with a Calcium-binding Protein (NEFA) in Neuronal Cytoplasm

Naoko Taniguchi; Hideo Taniura; Michio Niinobe; Chitoshi Takayama; Keiko Tominaga-Yoshino; Akihiko Ogura; Kazuaki Yoshikawa

Necdin, a growth suppressor expressed predominantly in postmitotic neurons, interacts with viral oncoproteins and cellular transcription factors E2F1 and p53. In search of other cellular targets of necdin, we screened cDNA libraries from neurally differentiated murine embryonal carcinoma P19 cells and adult rat brain by the yeast two-hybrid assay. We isolated cDNAs encoding partial sequences of mouse NEFA and rat nucleobindin (CALNUC), which are Ca2+-binding proteins possessing similar domain structures. Necdin interacted with NEFA via a domain encompassing two EF hand motifs, which had Ca2+ binding activity as determined by 45Ca2+ overlay. NEFA was widely distributed in mouse organs, whereas necdin was expressed predominantly in the brain and skeletal muscle. In mouse brain in vivo, NEFA was localized in neuronal perikarya and dendrites. By immunoelectron microscopy, NEFA was localized to the cisternae of the endoplasmic reticulum and nuclear envelope in brain neurons. NEFA-green fluorescent protein (GFP) fusion protein expressed in neuroblastoma N1E-115 cells was retained in the cytoplasm and partly secreted into the culture medium. Necdin enhanced the cytoplasmic retention of NEFA-GFP and potentiated the effect of NEFA-GFP on caffeine-evoked elevation of cytosolic Ca2+ levels. Thus, necdin and NEFA might be involved in Ca2+ homeostasis in neuronal cytoplasm.


Developmental Neuroscience | 2000

Cellular and Subcellular Localization of Necdin in Fetal and Adult Mouse Brain

Michio Niinobe; Kazuya Koyama; Kazuaki Yoshikawa

Necdin is a 325-amino-acid residue protein encoded by a cDNA clone isolated from neurally differentiated embryonal carcinoma cells. Ectopic expression of necdin induces growth arrest of proliferative cells. Necdin binds to major transcription factors E2F1 and p53, suggesting that necdin exerts its functions through the interactions with these cell-cycle-regulating factors. However, information about precise localization of endogenous necdin protein is currently lacking. A rabbit polyclonal antibody was raised against a bacterially expressed recombinant protein of necdin (amino acids 83–325). Immunoblot analysis revealed that necdin protein was expressed almost exclusively in the brain of adult mice. A relative molecular mass of endogenous necdin was estimated at approximately 43,000. In developing mouse brain, necdin was most abundant during fetal and neonatal periods. Necdin was highly enriched in the cytoplasm of hypothalamic neurons in fetal and adult mice. The subcellular fractionation analysis revealed that necdin was concentrated in the cytosol fraction of brain cells. These results suggest that endogenous necdin protein is localized predominantly in the cytoplasm of differentiated neurons and moves into the nucleus under specific conditions.


Gene | 1998

The human chromosomal gene for necdin, a neuronal growth suppressor, in the Prader–Willi syndrome deletion region

Yuji Nakada; Hideo Taniura; Taichi Uetsuki; Joji Inazawa; Kazuaki Yoshikawa

Necdin is a growth suppressor expressed in virtually all postmitotic neurons in the brain. The human necdin gene, NDN, is maternally imprinted and deleted in the Prader-Willi syndrome, a neurobehavioral contiguous gene disorder. Here, we isolated and characterized the human chromosomal necdin gene and its promoter region. The necdin gene is intronless, and it encodes a protein of 321 amino acid residues, four residues shorter than mouse Necdin. By fluorescence in-situ hybridization analysis, the necdin gene was localized to chromosome 15q11.2-q12 within the Prader-Willi syndrome deletion region. CpG islands were found in a region extending from the proximal 5-flanking sequence to the protein coding region. The 5-flanking sequence, which lacks canonical TATA and CAAT boxes, possessed a promoter activity in postmitotic neurons derived from murine embryonal carcinoma P19 cells. Methylation in vitro of HhaI CpG sites in the promoter region reduced the transcriptional activity. These results suggest that the necdin gene is silenced through methylation of the CpG island encompassing its promoter region.


Journal of Biological Chemistry | 2002

Ectopic expression of necdin induces differentiation of mouse neuroblastoma cells.

Masakatsu Kobayashi; Hideo Taniura; Kazuaki Yoshikawa

Necdin is expressed predominantly in postmitotic neurons, and ectopic expression of this protein strongly suppresses cell growth. Necdin has been implicated in the pathogenesis of Prader-Willi syndrome, a human neurodevelopmental disorder associated with genomic imprinting. Here we demonstrate that ectopic expression of necdin induces a neuronal phenotype in neuroblastoma cells. Necdin was undetectable in mouse neuroblastoma N1E-115 cells under undifferentiated and differentiated conditions. N1E-115 cells transfected with necdin cDNA showed morphological differentiation such as neurite outgrowth and expression of the synaptic marker proteins synaptotagmin and synaptophysin. In addition, Western blot analysis of the retinoblastoma protein (Rb) family members Rb, p130, and p107 revealed that necdin cDNA transfectants contained an increased level of p130 and a reduced level of p107, a pattern seen in differentiated G0 cells. The transcription factors E2F1 and E2F4 physically interacted with necdin via their carboxyl-terminal transactivation domains, but only E2F1 abrogated necdin-induced growth arrest and neurite outgrowth of neuroblastoma cells. Overexpression of E2F1 in differentiated N1E-115 cells induced apoptosis, which was antagonized by co-expression of necdin. These results suggest that necdin promotes the differentiation and survival of neurons through its antagonistic interactions with E2F1.

Collaboration


Dive into the Kazuaki Yoshikawa's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Takako Aizawa

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Kei Maruyama

Saitama Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge