Network


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

Hotspot


Dive into the research topics where Katsuhiko Asanuma is active.

Publication


Featured researches published by Katsuhiko Asanuma.


Nature Cell Biology | 2006

Synaptopodin orchestrates actin organization and cell motility via regulation of RhoA signalling

Katsuhiko Asanuma; Etsuko Yanagida-Asanuma; Christian Faul; Yasuhiko Tomino; Kwanghee Kim; Peter Mundel

The Rho family of small GTPases (RhoA, Rac1 and Cdc42) controls signal-transduction pathways that influence many aspects of cell behaviour, including cytoskeletal dynamics. At the leading edge, Rac1 and Cdc42 promote cell motility through the formation of lamellipodia and filopodia, respectively. On the contrary, RhoA promotes the formation of contractile actin–myosin-containing stress fibres in the cell body and at the rear. Here, we identify synaptopodin, an actin-associated protein, as a novel regulator of RhoA signalling and cell migration in kidney podocytes. We show that synaptopodin induces stress fibres by competitive blocking of Smurf1-mediated ubiquitination of RhoA, thereby preventing the targeting of RhoA for proteasomal degradation. Gene silencing of synaptopodin in kidney podocytes causes the loss of stress fibres and the formation of aberrant non-polarized filopodia and impairment of cell migration. Together, these data show that synaptopodin is essential for the integrity of the podocyte actin cytoskeleton and for the regulation of podocyte cell migration.


Journal of Clinical Investigation | 2005

Synaptopodin regulates the actin-bundling activity of α-actinin in an isoform-specific manner

Katsuhiko Asanuma; Kwanghee Kim; Jun Oh; Laura Giardino; Sophie Chabanis; Christian Faul; Jochen Reiser; Peter Mundel

Synaptopodin is the founding member of a novel class of proline-rich actin-associated proteins highly expressed in telencephalic dendrites and renal podocytes. Synaptopodin-deficient (synpo(-/-)) mice lack the dendritic spine apparatus and display impaired activity-dependent long-term synaptic plasticity. In contrast, the ultrastructure of podocytes in synpo(-/-) mice is normal. Here we show that synpo(-/-) mice display impaired recovery from protamine sulfate-induced podocyte foot process (FP) effacement and LPS-induced nephrotic syndrome. Similarly, synpo(-/-) podocytes show impaired actin filament reformation in vitro. We further demonstrate that synaptopodin exists in 3 isoforms, neuronal Synpo-short (685 AA), renal Synpo-long (903 AA), and Synpo-T (181 AA). The C terminus of Synpo-long is identical to that of Synpo-T. All 3 isoforms specifically interact with alpha-actinin and elongate alpha-actinin-induced actin filaments. synpo(-/-) mice lack Synpo-short and Synpo-long expression but show an upregulation of Synpo-T protein expression in podocytes, though not in the brain. Gene silencing of Synpo-T abrogates stress-fiber formation in synpo(-/-) podocytes, demonstrating that Synpo-T serves as a backup for Synpo-long in synpo(-/-) podocytes. In concert, synaptopodin regulates the actin-bundling activity of alpha-actinin in highly dynamic cell compartments, such as podocyte FPs and the dendritic spine apparatus.


The FASEB Journal | 2003

MAP-LC3, a promising autophagosomal marker, is processed during the differentiation and recovery of podocytes from PAN nephrosis

Katsuhiko Asanuma; Isei Tanida; Isao Shirato; Takashi Ueno; Hisatsugu Takahara; Tomohito Nishitani; Eiki Kominami; Yasuhiko Tomino

Microtubule‐associated protein 1 light chain 3 (LC3) is a unique modifier protein. LC3‐I, the cytosolic form, is modified to LC3‐II, the membrane‐bound form, by a mechanism similar to ubiquitylation by E1‐ and E2‐like enzymes, Apg7p and Apg3p, respectively. In the present study, we found that LC3‐I is processed to LC3‐II during the differentiation and recovery from puromycin aminonucleoside‐induced nephrosis of podocytes. LC3 is especially expressed in the podocytes of rat kidney as the membrane‐bound form LC3‐II. Biochemical analysis using a conditionally immortalized mouse podocyte clone (MPC) revealed that LC3‐I is processed to LC3‐II during the differentiation of cells into mature podocytes and accumulates in the membrane‐rich fraction of the cell lysate. LC3‐II‐localized vesicles, which differ from lysosomes and endosomes, in differentiated MPC cells are morphologically similar to autophagic vacuoles during starvation‐induced autophagy. During starvation‐induced autophagy, autophagosomes fuses with lysosome and LC3‐II on autophagosomes is finally degraded by lysosomal proteases. However, in differentiated MPC cells, little LC3‐II on the vesicles is degraded by lysosomal proteases, suggesting that little LC3‐II‐localized vesicles in differentiated MPC cells fuse with lysosome. Furthermore, the LC3‐II level in differentiated MPC cells increases with recovery from damage caused by experimental puromycin aminonucleoside‐induced nephrosis. These results suggest that LC3‐II‐localized vesicles play an important role in the physiological function of podocytes.


Journal of Clinical Investigation | 2006

Bigenic mouse models of focal segmental glomerulosclerosis involving pairwise interaction of CD2AP, Fyn, and synaptopodin

Tobias B. Huber; Christopher Kwoh; Hui Wu; Katsuhiko Asanuma; Markus Gödel; Björn Hartleben; Ken J. Blumer; Jeffrey H. Miner; Peter Mundel; Andrey S. Shaw

Focal segmental glomerulosclerosis (FSGS) is the most common primary glomerular diagnosis resulting in end-stage renal disease. Defects in several podocyte proteins have been implicated in the etiology of FSGS, including podocin, alpha-actinin-4, CD2-associated protein (CD2AP), and TRPC6. Despite our growing understanding of genes involved in the pathogenesis of focal segmental sclerosis, the vast majority of patients with this disease, even those with a familial linkage, lack a clear genetic diagnosis. Here, we tested whether combinations of genetic heterozygosity (bigenic heterozygosity) that alone do not result in clinical kidney disease could function together to enhance susceptibility to glomerular damage and FSGS. Combinations of Cd2ap heterozygosity and heterozygosity of either synaptopodin (Synpo) or Fyn proto-oncogene (Fyn) but not kin of IRRE like 1 (Neph1) resulted in spontaneous proteinuria and in FSGS-like glomerular damage. These genetic interactions were also reflected at a functional level, as we found that CD2AP associates with Fyn and Synpo but not with Neph1. This demonstrates that bigenic heterozygosity can lead to FSGS and suggests that combined mutations in 2 or multiple podocyte genes may be a common etiology for glomerular disease.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Nuclear relocation of the nephrin and CD2AP-binding protein dendrin promotes apoptosis of podocytes

Katsuhiko Asanuma; Kirk N. Campbell; Kwanghee Kim; Christian Faul; Peter Mundel

Kidney podocytes and their slit diaphragms (SDs) form the final barrier to urinary protein loss. There is mounting evidence that SD proteins also participate in intracellular signaling pathways. The SD protein nephrin serves as a component of a signaling complex that directly links podocyte junctional integrity to actin cytoskeletal dynamics. Another SD protein, CD2-associated protein (CD2AP), is an adaptor molecule involved in podocyte homeostasis that can repress proapoptotic TGF-β signaling in podocytes. Here we show that dendrin, a protein originally identified in telencephalic dendrites, is a constituent of the SD complex, where it directly binds to nephrin and CD2AP. In experimental glomerulonephritis, dendrin relocates from the SD to the nucleus of injured podocytes. High-dose, proapoptotic TGF-β1 directly promotes the nuclear import of dendrin, and nuclear dendrin enhances both staurosporine- and TGF-β1-mediated apoptosis. In summary, our results identify dendrin as an SD protein with proapoptotic signaling properties that accumulates in the podocyte nucleus in response to glomerular injury and provides a molecular target to tackle proteinuric kidney diseases. Nuclear relocation of dendrin may provide a mechanism whereby changes in SD integrity could translate into alterations of podocyte survival under pathological conditions.


Diabetes | 2016

Impaired podocyte autophagy exacerbates proteinuria in diabetic nephropathy

Atsuko Tagawa; Mako Yasuda; Shinji Kume; Kosuke Yamahara; Jun Nakazawa; Masami Chin-Kanasaki; Hisazumi Araki; Shin-ichi Araki; Daisuke Koya; Katsuhiko Asanuma; Eun-Hee Kim; Masakazu Haneda; Nobuyuki Kajiwara; Kazuyuki Hayashi; Hiroshi Ohashi; Satoshi Ugi; Hiroshi Maegawa; Takashi Uzu

Overcoming refractory massive proteinuria remains a clinical and research issue in diabetic nephropathy. This study was designed to investigate the pathogenesis of massive proteinuria in diabetic nephropathy, with a special focus on podocyte autophagy, a system of intracellular degradation that maintains cell and organelle homeostasis, using human tissue samples and animal models. Insufficient podocyte autophagy was observed histologically in patients and rats with diabetes and massive proteinuria accompanied by podocyte loss, but not in those with no or minimal proteinuria. Podocyte-specific autophagy-deficient mice developed podocyte loss and massive proteinuria in a high-fat diet (HFD)–induced diabetic model for inducing minimal proteinuria. Interestingly, huge damaged lysosomes were found in the podocytes of diabetic rats with massive proteinuria and HFD-fed, podocyte-specific autophagy-deficient mice. Furthermore, stimulation of cultured podocytes with sera from patients and rats with diabetes and massive proteinuria impaired autophagy, resulting in lysosome dysfunction and apoptosis. These results suggest that autophagy plays a pivotal role in maintaining lysosome homeostasis in podocytes under diabetic conditions, and that its impairment is involved in the pathogenesis of podocyte loss, leading to massive proteinuria in diabetic nephropathy. These results may contribute to the development of a new therapeutic strategy for advanced diabetic nephropathy.


Nephrology | 2007

The role of podocytes in proteinuria.

Katsuhiko Asanuma; Etsuko Yanagida-Asanuma; Miyuki Takagi; Fumiko Kodama; Yasuhiko Tomino

SUMMARY:  Glomerular visceral epithelial cells, also known as podocytes, are highly specialized epithelial cells that cover the outer layer of the glomerular basement membrane. Podocytes consist of cell bodies, major processes and foot processes (FP) of neighbouring cells, with the filtration slits bridged by the slit membrane between them. The function of podocytes is largely based on their specialized cell architecture and functions such as stabilization of glomerular capillaries and participation in the barrier function of the glomerular filter. Therefore, they form the final barrier to protein loss, which explains why podocyte injury is typically associated with marked proteinuria. Under pathological conditions, podocytes exhibit various changes. Among these changes, FP effacement represents the most characteristic change in cell shape of podocytes. FP effacement is dependent on disruption of the actin cytoskeletal network in the podocytes, The mechanisms of organization and re‐organization of actin in the FP of podocytes are discussed in this review.


Laboratory Investigation | 2011

mTORC1 activation triggers the unfolded protein response in podocytes and leads to nephrotic syndrome.

Noriko Ito; Yukino Nishibori; Yugo Ito; Hisashi Takagi; Yoshihiro Akimoto; Akihiko Kudo; Katsuhiko Asanuma; Yoshimichi Sai; Ken-ichi Miyamoto; Hitoshi Takenaka; Kunimasa Yan

Although podocyte damage is known to be responsible for the development of minimal-change disease (MCD), the underlying mechanism remains to be elucidated. Previously, using a rat MCD model, we showed that endoplasmic reticulum (ER) stress in the podocytes was associated with the heavy proteinuric state and another group reported that a mammalian target of rapamycin complex 1 (mTORC1) inhibitor protected against proteinuria. In this study, which utilized a rat MCD model, a combination of immunohistochemistry, dual immunofluorescence and confocal microscopy, western blot analysis, and quantitative real-time RT-PCR revealed co-activation of the unfolded protein response (UPR), which was induced by ER stress, and mTORC1 in glomerular podocytes before the onset of proteinuria and downregulation of nephrin at the post-translational level at the onset of proteinuria. Podocyte culture experiments revealed that mTORC1 activation preceded the UPR that was associated with a marked decrease in the energy charge. The mTORC1 inhibitor everolimus completely inhibited proteinuria through a reduction in both mTORC1 and UPR activity and preserved nephrin expression in the glomerular podocytes. In conclusion, mTORC1 activation may perturb the regulatory system of energy metabolism primarily by promoting energy consumption and inducing the UPR, which underlie proteinuria in MCD.


Clinical Journal of The American Society of Nephrology | 2012

Relationships between Levels of Urinary Podocalyxin, Number of Urinary Podocytes, and Histologic Injury in Adult Patients with IgA Nephropathy

Rin Asao; Katsuhiko Asanuma; Fumiko Kodama; Miyuki Akiba-Takagi; Yoshiko Nagai-Hosoe; Takuto Seki; Yukihiko Takeda; Isao Ohsawa; Satoshi Mano; Kiyoshi Matsuoka; Hiroyuki Kurosawa; Shinya Ogasawara; Sakari Sekine; Satoshi Horikoshi; Masanori Hara; Yasuhiko Tomino

BACKGROUND AND OBJECTIVES Podocalyxin (PCX) is present on the apical cell membrane of podocytes and is shed in urine from injured podocytes. Urinary podocalyxin (u-PCX) is associated with severity of active glomerular injury in patients with glomerular diseases. This study examined the relationship between number of urinary podocytes, levels of u-PCX, and glomerular injury in adults with IgA nephropathy (IgAN). DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Urine samples voided in the morning on the day of biopsy were obtained from 51 patients with IgAN (18 men and 33 women; mean age, 31 years). All renal biopsy specimens were analyzed histologically. Pathologic variables of IgAN were analyzed per Shigematsu classification, the Oxford classification of IgAN, and the Clinical Guidelines of IgAN in Japan. Levels of u-PCX were measured by sandwich ELISA. RESULTS Histologic analysis based on Shigematsu classification revealed a significant correlation between levels of u-PCX and severity of acute extracapillary abnormalities (r=0.72; P<0.001), but levels of urinary protein excretion did not correlate with acute glomerular abnormalities. Levels of urinary protein excretion in patients with segmental sclerosis (n=19) were higher than in patients without (n=22) (0.49 [interquartile range (IQR), 0.20-0.88] g/g creatinine versus 0.20 [IQR, 0.10-0.33] g/g creatinine; P<0.01). The number of urinary podocytes in patients with segmental sclerosis was higher than in patients without (1.05 [IQR, 0.41-1.67] per mg creatinine versus 0.28 [IQR, 0.10-0.66] per mg creatinine; P<0.01). CONCLUSIONS Levels of u-PCX and the number of urinary podocytes are associated with histologic abnormalities in adults with IgAN.


Journal of Hypertension | 2010

Cilnidipine suppresses podocyte injury and proteinuria in metabolic syndrome rats: possible involvement of N-type calcium channel in podocyte

Yu-Yan Fan; Masakazu Kohno; Daisuke Nakano; Hiroyuki Ohsaki; Hiroyuki Kobori; Diah Suwarni; Naro Ohashi; Hirofumi Hitomi; Katsuhiko Asanuma; Takahisa Noma; Yasuhiko Tomino; Toshiro Fujita; Akira Nishiyama

Objectives Clinical studies have indicated the beneficial effect of an L/N-type calcium channel blocker (CCB), cilnidipine, on the progression of proteinuria in hypertensive patients compared with an L-type CCB, amlodipine. In the present study, we examined the effects of cilnidipine and amlodipine on the renal injury in spontaneously hypertensive rat/ND mcr-cp (SHR/ND) and their underlying mechanism. Methods and results SHR/ND were treated with vehicle (n = 10), cilnidipine [33 mg/kg per day, orally (p.o.); n = 11] or amlodipine (20 mg/kg per day, p.o.; n = 9) for 20 weeks. SHR/ND developed proteinuria in an age-dependent manner. Cilnidipine suppressed the proteinuria greater than amlodipine did. The immunohistochemical analysis showed that N-type calcium channel and Wilms tumor factor, a marker of podocyte, were co-expressed. SHR/ND had significantly greater desmin staining, an indicator of podocyte injury, with lower podocin and nephrin expression in the glomeruli than Wistar–Kyoto rat or SHR. Cilnidipine significantly prevented the increase in desmin staining and restored the glomerular podocin and nephrin expression compared with amlodipine. Cilnidipine also prevented the increase in renal angiotensin II content, the expression and membrane translocation of NADPH oxidase subunits and dihydroethidium staining in SHR/ND. In contrast, amlodipine failed to change these renal parameters. Conclusion These data suggest that cilnidipine suppressed the development of proteinuria greater than amlodipine possibly through inhibiting N-type calcium channel-dependent podocyte injury in SHR/ND.

Collaboration


Dive into the Katsuhiko Asanuma's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge