Network


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

Hotspot


Dive into the research topics where Yumiko Azuma is active.

Publication


Featured researches published by Yumiko Azuma.


PLOS ONE | 2012

Knockdown of the Drosophila fused in sarcoma (FUS) homologue causes deficient locomotive behavior and shortening of motoneuron terminal branches.

Hiroshi Sasayama; Mai Shimamura; Takahiko Tokuda; Yumiko Azuma; Tomokatsu Yoshida; Toshiki Mizuno; Masanori Nakagawa; Nobuhiro Fujikake; Yoshitaka Nagai; Masamitsu Yamaguchi

Mutations in the fused in sarcoma/translated in liposarcoma gene (FUS/TLS, FUS) have been identified in sporadic and familial forms of amyotrophic lateral sclerosis (ALS). FUS is an RNA-binding protein that is normally localized in the nucleus, but is mislocalized to the cytoplasm in ALS, and comprises cytoplasmic inclusions in ALS-affected areas. However, it is still unknown whether the neurodegeneration that occurs in ALS is caused by the loss of FUS nuclear function, or by the gain of toxic function due to cytoplasmic FUS aggregation. Cabeza (Caz) is a Drosophila orthologue of human FUS. Here, we generated Drosophila models with Caz knockdown, and investigated their phenotypes. In wild-type Drosophila, Caz was strongly expressed in the central nervous system of larvae and adults. Caz did not colocalize with a presynaptic marker, suggesting that Caz physiologically functions in neuronal cell bodies and/or their axons. Fly models with neuron-specific Caz knockdown exhibited reduced climbing ability in adulthood and anatomical defects in presynaptic terminals of motoneurons in third instar larvae. Our results demonstrated that decreased expression of Drosophila Caz is sufficient to cause degeneration of motoneurons and locomotive disability in the absence of abnormal cytoplasmic Caz aggregates, suggesting that the pathogenic mechanism underlying FUS-related ALS should be ascribed more to the loss of physiological FUS functions in the nucleus than to the toxicity of cytoplasmic FUS aggregates. Since the Caz-knockdown Drosophila model we presented recapitulates key features of human ALS, it would be a suitable animal model for the screening of genes and chemicals that might modify the pathogenic processes that lead to the degeneration of motoneurons in ALS.


Human Molecular Genetics | 2014

Identification of ter94, Drosophila VCP, as a strong modulator of motor neuron degeneration induced by knockdown of Caz, Drosophila FUS

Yumiko Azuma; Takahiko Tokuda; Mai Shimamura; Akane Kyotani; Hiroshi Sasayama; Tomokatsu Yoshida; Ikuko Mizuta; Toshiki Mizuno; Masanori Nakagawa; Nobuhiro Fujikake; Morio Ueyama; Yoshitaka Nagai; Masamitsu Yamaguchi

In humans, mutations in the fused in sarcoma (FUS) gene have been identified in sporadic and familial forms of amyotrophic lateral sclerosis (ALS). Cabeza (Caz) is the Drosophila ortholog of human FUS. Previously, we established Drosophila models of ALS harboring Caz-knockdown. These flies develop locomotive deficits and anatomical defects in motoneurons (MNs) at neuromuscular junctions; these phenotypes indicate that loss of physiological FUS functions in the nucleus can cause MN degeneration similar to that seen in FUS-related ALS. Here, we aimed to explore molecules that affect these ALS-like phenotypes of our Drosophila models with eye-specific and neuron-specific Caz-knockdown. We examined several previously reported ALS-related genes and found genetic links between Caz and ter94, the Drosophila ortholog of human Valosin-containing protein (VCP). Genetic crossing the strongest loss-of-function allele of ter94 with Caz-knockdown strongly enhanced the rough-eye phenotype and the MN-degeneration phenotype caused by Caz-knockdown. Conversely, the overexpression of wild-type ter94 in the background of Caz-knockdown remarkably suppressed those phenotypes. Our data demonstrated that expression levels of Drosophila VCP ortholog dramatically modified the phenotypes caused by Caz-knockdown in either direction, exacerbation or remission. Our results indicate that therapeutic agents that up-regulate the function of human VCP could modify the pathogenic processes that lead to the degeneration of MNs in ALS.


Experimental Neurology | 2016

Knockdown of the Drosophila FIG4 induces deficient locomotive behavior, shortening of motor neuron, axonal targeting aberration, reduction of life span and defects in eye development

Akane Kyotani; Yumiko Azuma; I. Yamamoto; Hideki Yoshida; Ikuko Mizuta; Toshiki Mizuno; Masanori Nakagawa; Takahiko Tokuda; Masamitsu Yamaguchi

Mutations in Factor-Induced-Gene 4 (FIG4) gene have been identified in Charcot-Marie-Tooth disease type 4J (CMT4J), Yunis-Varon syndrome and epilepsy with polymicrogyria. FIG4 protein regulates a cellular abundance of phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2), a signaling lipid on the cytosolic surface of membranes of the late endosomal compartment. PI(3,5)P2 is required for retrograde membrane trafficking from lysosomal and late endosomal compartments to the Golgi. However, it is still unknown how the neurodegeneration that occurs in these diseases is related to the loss of FIG4 function. Drosophila has CG17840 (dFIG4) as a human FIG4 homolog. Here we specifically knocked down dFIG4 in various tissues, and investigated their phenotypes. Neuron-specific knockdown of dFIG4 resulted in axonal targeting aberrations of photoreceptor neurons, shortened presynaptic terminals of motor neurons in 3rd instar larvae and reduced climbing ability in adulthood and life span. Fat body-specific knockdown of dFIG4 resulted in enlarged lysosomes in cells that were detected by staining with LysoTracker. In addition, eye imaginal disk-specific knockdown of dFIG4 disrupted differentiation of pupal ommatidial cell types, such as cone cells and pigment cells, suggesting an additional role of dFIG4 during eye development.


Journal of Endovascular Therapy | 2007

Tandem Balloon Protection during Emergency Carotid Artery Stenting of a Stenotic ICA Lesion with Intraluminal Thrombus

Keisuke Imai; Masahiro Makino; Fumitoshi Niwa; Tetsuro Takegami; Hironori Koike; Yumiko Azuma; Fumiko Oshima; Kenichiro Oda; Kiyohito Kakita

Purpose: To report a technique for emergency CAS of a stenotic internal carotid artery (ICA) lesion with an intraluminal thrombus, which predisposes to distal thromboembolism and could aggravate pre-existing neurological symptoms. Case Report: A 77-year-old man with fluctuating stroke underwent successful emergency CAS for an ICA stenosis with intraluminal thrombus using a tandem balloon protection technique featuring proximal balloon blockade and a PercuSurge GuardWire system for distal protection. Conclusion: This experience suggests that emergency CAS with tandem balloon protection is a potential treatment for a stenotic lesion with an intraluminal thrombus in patients with fluctuating stroke.


Experimental Cell Research | 2014

Genetic link between Cabeza, a Drosophila homologue of Fused in Sarcoma (FUS), and the EGFR signaling pathway.

Mai Shimamura; Akane Kyotani; Yumiko Azuma; Hideki Yoshida; Thanh Binh Nguyen; Ikuko Mizuta; Tomokatsu Yoshida; Toshiki Mizuno; Masanori Nakagawa; Takahiko Tokuda; Masamitsu Yamaguchi

Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease that causes progressive muscular weakness. Fused in Sarcoma (FUS) that has been identified in familial ALS is an RNA binding protein that is normally localized in the nucleus. However, its function in vivo is not fully understood. Drosophila has Cabeza (Caz) as a FUS homologue and specific knockdown of Caz in the eye imaginal disc and pupal retina using a GMR-GAL4 driver was here found to induce an abnormal morphology of the adult compound eyes, a rough eye phenotype. This was partially suppressed by expression of the apoptosis inhibitor P35. Knockdown of Caz exerted no apparent effect on differentiation of photoreceptor cells. However, immunostaining with an antibody to Cut that marks cone cells revealed fusion of these and ommatidia of pupal retinae. These results indicate that Caz knockdown induces apoptosis and also inhibits differentiation of cone cells, resulting in abnormal eye morphology in adults. Mutation in EGFR pathway-related genes, such as rhomboid-1, rhomboid-3 and mirror suppressed the rough eye phenotype induced by Caz knockdown. Moreover, the rhomboid-1 mutation rescued the fusion of cone cells and ommatidia observed in Caz knockdown flies. The results suggest that Caz negatively regulates the EGFR signaling pathway required for determination of cone cell fate in Drosophila.


Journal of Clinical Neuromuscular Disease | 2014

Sixty six-month follow-up of muscle power and respiratory function in a case with adult-type Pompe disease treated with enzyme replacement therapy.

Reina Isayama; Kensuke Shiga; Kazuya Seo; Yumiko Azuma; Yasukiyo Araki; Ai Hamano; Hedesato Takezawa; Nagato Kuriyama; Nobuo Takezawa; Toshiki Mizuno; Masanori Nakagawa

Abstract We report a patient with adult-type Pompe disease treated with enzyme replacement therapy (ERT) for 5.5 years. We evaluated pulmonary function and muscle strength using 6-minute walk test, manual muscle test, and dynamometer-based measurement. The long-term ERT resulted in a substantial improvement in the pulmonary function and a possible stabilization followed by mild deterioration in muscle power measured by dynamometer and 6-minute walk test. Our data may rationalize the long-term use of ERT for adult-type Pompe disease in terms of maintaining pulmonary function.


Auris Nasus Larynx | 2011

A case report of HTLV-I associated myelopathy presenting with cerebellar ataxia and nystagmus.

Masakatsu Taki; Fumiaki Nin; Tatsuhisa Hasegawa; Hirofumi Sakaguchi; Toshihiro Suzuki; Yasuo Hisa; Yumiko Azuma; Masanori Nakagawa

HTLV-I associated myelopathy/tropical spastic paraparesis (HAM/TSP) is characterized by spastic paraparesis in the lower extremities, and urinary disturbance. HAM/TSP has also been less frequently associated with cerebellar syndromes and nystagmus. We report a case of HAM/TSP presenting with cerebellar ataxia and nystagmus. The patient was a 73-year-old woman who was born in southern Japan. At age 41, she developed pain and spasticity in the bilateral lower limbs and gradually progressive gait disturbance. At age 57, she was diagnosed with HAM/TSP based on spastic paraparesis in the lower limbs, urinary disturbance and positive anti HTLV-I antibody in serum and cerebrospinal fluid. In June 2008, she was referred to our university and hospitalized for rehabilitation. Twenty days later, she experienced rotatory vertigo sensation. Magnetic resonance imaging revealed pontocerebellar atrophy. The patient presented with cerebellar signs in the upper limbs, gaze-evoked nystagmus in the sitting position and right-beating horizontal nystagmus in the supine and head-hanging positions. Electronystagmography (ENG) showed horizontal saccadic overshoot dysmetria and horizontal saccadic pursuit. Nystagmus is rare among the literature on HAM/TSP. ENG is helpful to evaluate and confirm the cerebellar syndromes of HAM/TSP.


npj Parkinson's disease | 2017

Early-onset parkinsonism in a pedigree with phosphoglycerate kinase deficiency and a heterozygous carrier: do PGK-1 mutations contribute to vulnerability to parkinsonism?

Satoshi Sakaue; Takashi Kasai; Ikuko Mizuta; Masaya Suematsu; Shinya Osone; Yumiko Azuma; Toshihiko Imamura; Takahiko Tokuda; Hitoshi Kanno; Omar M. A. El-Agnaf; Masafumi Morimoto; Masanori Nakagawa; Hajime Hosoi; Toshiki Mizuno

Phosphoglycerate kinase 1 (PGK-1) is a glycolytic enzyme encoded by PGK-1, which maps to the X chromosome. PGK-1 deficiency causes X-linked recessive hereditary chronic hemolytic anemia, myopathy, and neurological disorders due to insufficient ATP regeneration. Early-onset parkinsonism has occasionally been reported as a neurological complication of this condition. However, heterozygous carriers of PGK-1 deficiency were thought to be neurologically asymptomatic. Here, we report a boy with PGK-1 deficiency and his mother, a carrier of a heterozygous mutation in PGK-1, both of whom presented with early-onset parkinsonism. The boy developed parkinsonism at 9 years of age. His parkinsonism partially responded to levodopa treatment. 123l-metaiodobenzylguanidine (MIBG) uptake was normal. His mother, who exhibited normal PGK-1 activity in erythrocytes, developed parkinsonism at 36 years of age. Her symptoms were undistinguishable from those of Parkinson’s disease (PD), despite her normal uptake of MIBG. Neither a point mutation in nor multiplication of SNCA was found. Additionally, hotspots of LRRK2 and GBA were not mutated. To our knowledge, this report provides the first description of parkinsonism in a carrier of PGK-1 deficiency. Interestingly, PGK-1 is located within the confirmed susceptibility locus for PD known as PARK12. These observations suggest that PGK-1 mutations confer susceptibility to PD.Early-onset Parkinson’s disease: a new genetic linkMutations in the gene encoding phosphoglycerate kinase 1 (PGK-1) may confer susceptibility to early-onset Parkinson’s disease (PD). PGK-1 is a crucial protein for the breakdown of sugar in the body and mutations that cause PGK-1 deficiency lead to an X-linked metabolic disorder characterised by the breakdown of red blood cells, muscle weakness and various central nervous system abnormalities. Takashi Kasai at Kyoto Prefectural University of Medicine, Japan, and colleagues describe early-onset PD symptoms in a 9 year old boy with PGK-1 deficiency and his mother at 36 years of age. This is the first report of parkinsonism developing in an otherwise asymptomatic carrier of a PGK-1 mutation. The location of the PGK-1 gene on the X chromosome is within a confirmed susceptibility region for PD known as PARK12, suggesting that PGK-1 may directly contribute to the disease.


Scientific Reports | 2018

NPM-hMLF1 fusion protein suppresses defects of a Drosophila FTLD model expressing the human FUS gene

I. Yamamoto; Yumiko Azuma; Yukie Kushimura; Hideki Yoshida; Ikuko Mizuta; Toshiki Mizuno; Morio Ueyama; Yoshitaka Nagai; Takahiko Tokuda; Masamitsu Yamaguchi

Fused in sarcoma (FUS) was identified as a component of typical inclusions in frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). In FTLD, both nuclear and cytoplasmic inclusions with wild-type FUS exist, while cytoplasmic inclusions with a mutant-form of FUS occur in many ALS cases. These observations imply that FUS plays a role across these two diseases. In this study, we examined the effect of several proteins including molecular chaperons on the aberrant eye morphology phenotype induced by overexpression of wild-type human FUS (hFUS) in Drosophila eye imaginal discs. By screening, we found that the co-expression of nucleophosmin–human myeloid leukemia factor 1 (NPM-hMLF1) fusion protein could suppress the aberrant eye morphology phenotype induced by hFUS. The driving of hFUS expression at 28 °C down-regulated levels of hFUS and endogenous cabeza, a Drosophila homolog of hFUS. The down-regulation was mediated by proteasome dependent degradation. Co-expression of NPM-hMLF1 suppressed this down-regulation. In addition, co-expression of NPM-hMLF1 partially rescued pharate adult lethal phenotype induced by hFUS in motor neurons. These findings with a Drosophila model that mimics FTLD provide clues for the development of novel FTLD therapies.


Experimental Neurology | 2018

Genetic screening of the genes interacting with Drosophila FIG4 identified a novel link between CMT-causing gene and long noncoding RNAs

Yuuka Muraoka; Aya Nakamura; Ryo Tanaka; Kojiro Suda; Yumiko Azuma; Yukie Kushimura; Luca Lo Piccolo; Hideki Yoshida; Ikuko Mizuta; Takahiko Tokuda; Toshiki Mizuno; Masanori Nakagawa; Masamitsu Yamaguchi

ABSTRACT Neuron‐specific knockdown of the dFIG4 gene, a Drosophila homologue of human FIG4 and one of the causative genes for Charcot‐Marie‐Tooth disease (CMT), reduces the locomotive abilities of adult flies, as well as causing defects at neuromuscular junctions, such as reduced synaptic branch length in presynaptic terminals of the motor neurons in third instar larvae. Eye imaginal disc‐specific knockdown of dFIG4 induces abnormal morphology of the adult compound eye, the rough eye phenotype. In this study, we carried out modifier screening of the dFIG4 knockdown‐induced rough eye phenotype using a set of chromosomal deficiency lines on the second chromosome. By genetic screening, we detected 9 and 15 chromosomal regions whose deletions either suppressed or enhanced the rough eye phenotype induced by the dFIG4 knockdown. By further genetic screening with mutants of individual genes in one of these chromosomal regions, we identified the gene CR18854 that suppressed the rough eye phenotype and the loss‐of‐cone cell phenotype. The CR18854 gene encodes a long non‐coding RNA (lncRNA) consisting of 2566 bases. Mutation and knockdown of CR18854 patially suppressed the enlarged lysosome phenotype induced by Fat body‐specific knockdown of dFIG4. Further characterization of CR18854, and a few other lncRNAs in relation to dFIG4 in neuron, using neuron‐specific dFIG4 knockdown flies indicated a genetic link between the dFIG4 gene and lncRNAs including CR18854 and hsr&ohgr;. We also obtained data indicating genetic interaction between CR18854 and Cabeza, a Drosophila homologue of human FUS, which is one of the causing genes for amyotrophic lateral sclerosis (ALS). These results suggest that lncRNAs such as CR18854 and hsr&ohgr; are involved in a common pathway in CMT and ALS pathogenesis. HIGHLIGHTSThe dFIG4 gene, a Drosophila homologue of human FIG4 is a causing genes for Charcot‐Marie‐Tooth disease.The CR18854 gene encoding a long hairpin RNA was identified as a genetic interactant with dFIG4.The other long non‐coding RNA hsr&ohgr; also genetically interacts with dsss4.The CR18854 gene genetically interacts with Drosophila FUS (Cabeza), a causing gene for ALS.The lncRNAs such as CR18854 and hsr&ohgr; may involve in common pathway for CMT and ALS pathogenesis.

Collaboration


Dive into the Yumiko Azuma's collaboration.

Top Co-Authors

Avatar

Masamitsu Yamaguchi

Kyoto Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Toshiki Mizuno

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hideki Yoshida

Kyoto Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ikuko Mizuta

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

I. Yamamoto

Kyoto Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yukie Kushimura

Kyoto Prefectural University of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge