Network


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

Hotspot


Dive into the research topics where Daisuke Koya is active.

Publication


Featured researches published by Daisuke Koya.


Journal of Clinical Investigation | 1997

Characterization of protein kinase C beta isoform activation on the gene expression of transforming growth factor-beta, extracellular matrix components, and prostanoids in the glomeruli of diabetic rats.

Daisuke Koya; Michael R. Jirousek; You-Wei Lin; Hidehiro Ishii; Koji Kuboki; George L. King

Induction of protein kinase C (PKC) pathway in the vascular tissues by hyperglycemia has been associated with many of the cellular changes observed in the complications of diabetes. Recently, we have reported that the use of a novel, orally effective specific inhibitor of PKC beta isoform (LY333531) normalized many of the early retinal and renal hemodynamics in rat models of diabetes. In the present study, we have characterized a spectrum of biochemical and molecular abnormalities associated with chronic changes induced by glucose or diabetes in the cultured mesangial cells and renal glomeruli that can be prevented by LY333531. Hyperglycemia increased diacylglycerol (DAG) level in cultured mesangial cells exposed to high concentrations of glucose and activated PKC alpha and beta1 isoforms in the renal glomeruli of diabetic rats. The addition of PKC beta selective inhibitor (LY333531) to cultured mesangial cells inhibited activated PKC activities by high glucose without lowering DAG levels and LY333531 given orally in diabetic rats specifically inhibited the activation of PKC beta1 isoform without decreasing PKC alpha isoform activation. Glucose-induced increases in arachidonic acid release, prostaglandin E2 production, and inhibition of Na+-K+ ATPase activities in the cultured mesangial cells were completely prevented by the addition of LY333531. Oral feeding of LY333531 prevented the increased mRNA expression of TGF-beta1 and extracellular matrix components such as fibronectin and alpha1(IV) collagen in the glomeruli of diabetic rats in parallel with inhibition of glomerular PKC activity. These results suggest that the activation of PKC, predominately the beta isoform by hyperglycemia in the mesangial cells and glomeruli can partly contribute to early renal dysfunctions by alteration of prostaglandin production and Na+-K+ ATPase activity as well as the chronic pathological changes by the overexpression of TGF-beta1 and extracellular matrix components genes.


The FASEB Journal | 2000

Amelioration of accelerated diabetic mesangial expansion by treatment with a PKC β inhibitor in diabetic db/db mice, a rodent model for type 2 diabetes

Daisuke Koya; Masakazu Haneda; Hiroko Nakagawa; Keiji Isshiki; Haruhisa Sato; Shiro Maeda; Toshiro Sugimoto; Hitoshi Yasuda; Atsunori Kashiwagi; D. Kirk Ways; George L. King; Ryuichi Kikkawa

Activation of protein kinase C (PKC) is implicated as an important mechanism by which diabetes causes vascular complications. We have recently shown that a PKC β inhibitor ameliorates not only early diabetes‐induced glomerular dysfunction such as glomerular hyperfiltration and albuminuria, but also overexpression of glomerular mRNA for transforming growth factor β1 (TGF‐βΙ) and extracellular matrix (ECM) proteins in streptozoto‐cin‐induced diabetic rats, a model for type 1 diabetes. In this study, we examined the long‐term effects of a PKC β inhibitor on glomerular histology as well as on biochemical and functional abnormalities in glomeruli of db/db mice, a model for type 2 diabetes. Administration of a PKC β inhibitor reduced urinary albumin excretion rates and inhibited glo‐merular PKC activation in diabetic db/db mice. Administration of a PKC β inhibitor also prevented the mesangial expansion observed in diabetic db/db mice, possibly through attenuation of glomerular expression of TGF‐β and ECM proteins such as fibronectin and type IV collagen. These findings provide the first in vivo evidence that the long‐term inhibition of PKC activation in the renal glomeruli can ameliorate glomerular pathologies in diabetic state, and thus suggest that a PKC β inhibitor might be an useful therapeutic strategy for the treatment of diabetic nephropathy.—Koya, D., Haneda, M., Nakagawa, H., Isshiki, K., Sato, H., Maeda, S., Sugimoto, T., Yasuda, H., Kashiwagi, A., Ways, D. K., King, G. L., Kikkawa, R. Amelioration of accelerated diabetic mesangial expansion by treatment with A PKC b inhibitor in diabetic db/db mice, a rodent model for type 2 diabetes. FASEB J. 14, 439–447 (2000)


Journal of Clinical Investigation | 2010

Calorie restriction enhances cell adaptation to hypoxia through Sirt1-dependent mitochondrial autophagy in mouse aged kidney

Shinji Kume; Takashi Uzu; Kihachiro Horiike; Masami Chin-Kanasaki; Keiji Isshiki; Shin-ichi Araki; Toshiro Sugimoto; Masakazu Haneda; Atsunori Kashiwagi; Daisuke Koya

Mitochondrial oxidative damage is a basic mechanism of aging, and multiple studies demonstrate that this process is attenuated by calorie restriction (CR). However, the molecular mechanism that underlies the beneficial effect of CR on mitochondrial dysfunction is unclear. Here, we investigated in mice the mechanisms underlying CR-mediated protection against hypoxia in aged kidney, with a special focus on the role of the NAD-dependent deacetylase sirtuin 1 (Sirt1), which is linked to CR-related longevity in model organisms, on mitochondrial autophagy. Adult-onset and long-term CR in mice promoted increased Sirt1 expression in aged kidney and attenuated hypoxia-associated mitochondrial and renal damage by enhancing BCL2/adenovirus E1B 19-kDa interacting protein 3-dependent (Bnip3-dependent) autophagy. Culture of primary renal proximal tubular cells (PTCs) in serum from CR mice promoted Sirt1-mediated forkhead box O3 (Foxo3) deacetylation. This activity was essential for expression of Bnip3 and p27Kip1 and for subsequent autophagy and cell survival of PTCs under hypoxia. Furthermore, the kidneys of aged Sirt1+/- mice were resistant to CR-mediated improvement in the accumulation of damaged mitochondria under hypoxia. These data highlight the role of the Sirt1-Foxo3 axis in cellular adaptation to hypoxia, delineate a molecular mechanism of the CR-mediated antiaging effect, and could potentially direct the design of new therapies for age- and hypoxia-related tissue damage.


Nature Reviews Endocrinology | 2009

SIRT1 and insulin resistance.

Fengxia Liang; Shinji Kume; Daisuke Koya

Sirtuin 1 (SIRT1), the mammalian homolog of SIR2, was originally identified as a NAD-dependent histone deacetylase, the activity of which is closely associated with lifespan under calorie restriction. Growing evidence suggests that SIRT1 regulates glucose or lipid metabolism through its deacetylase activity for over two dozen known substrates, and has a positive role in the metabolic pathway through its direct or indirect involvement in insulin signaling. SIRT1 stimulates a glucose-dependent insulin secretion from pancreatic β cells, and directly stimulates insulin signaling pathways in insulin-sensitive organs. Furthermore, SIRT1 regulates adiponectin secretion, inflammatory responses, gluconeogenesis, and levels of reactive oxygen species, which together contribute to the development of insulin resistance. Moreover, overexpression of SIRT1 and several SIRT1 activators has beneficial effects on glucose homeostasis and insulin sensitivity in obese mice models. These findings suggest that SIRT1 might be a new therapeutic target for the prevention of disease related to insulin resistance, such as metabolic syndrome and diabetes mellitus, although direct evidence from clinical studies in humans is needed to prove this possibility. In this Review, we discuss the potential role and therapeutic promise of SIRT1 in insulin resistance on the basis of the latest experimental studies.


Journal of Biological Chemistry | 2007

SIRT1 inhibits transforming growth factor beta-induced apoptosis in glomerular mesangial cells via Smad7 deacetylation.

Shinji Kume; Masakazu Haneda; Keizo Kanasaki; Toshiro Sugimoto; Shin-ichi Araki; Keiji Isshiki; Motohide Isono; Takashi Uzu; Leonard Guarente; Atsunori Kashiwagi; Daisuke Koya

SIRT1, a class III histone deacetylase, is considered a key regulator of cell survival and apoptosis through its interaction with nuclear proteins. In this study, we have examined the likelihood and role of the interaction between SIRT1 and Smad7, which mediates transforming growth factor β (TGFβ)-induced apoptosis in renal glomerular mesangial cells. Immunoprecipitation analysis revealed that SIRT1 directly interacts with the N terminus of Smad7. Furthermore, SIRT1 reversed acetyl-transferase (p300)-mediated acetylation of two lysine residues (Lys-64 and -70) on Smad7. In mesangial cells, the Smad7 expression level was reduced by SIRT1 overexpression and increased by SIRT1 knockdown. SIRT1-mediated deacetylation of Smad7 enhanced Smad ubiquitination regulatory factor 1 (Smurf1)-mediated ubiquitin proteasome degradation, which contributed to the low expression of Smad7 in SIRT1-overexpressing mesangial cells. Stimulation by TGFβ or overexpression of Smad7 induced mesangial cell apoptosis, as assessed by morphological apoptotic changes (nuclear condensation) and biological apoptotic markers (cleavages of caspase3 and poly(ADP-ribose) polymerase). However, TGFβ failed to induce apoptosis in Smad7 knockdown mesangial cells, indicating that Smad7 mainly mediates TGFβ-induced apoptosis of mesangial cells. Finally, SIRT1 overexpression attenuated both Smad7- and TGFβ-induced mesangial cell apoptosis, whereas SIRT1 knockdown enhanced this apoptosis. We have concluded that Smad7 is a new target molecule for SIRT1 and SIRT1 attenuates TGFβ-induced mesangial cell apoptosis through acceleration of Smad7 degradation. Our results suggest that up-regulation of SIRT1 deacetylase activity is a potentially useful therapeutic strategy for prevention of TGFβ-related kidney disease through its effect on cell survival.


Journal of The American Society of Nephrology | 2007

Role of Altered Renal Lipid Metabolism in the Development of Renal Injury Induced by a High-Fat Diet

Shinji Kume; Takashi Uzu; Shin-ichi Araki; Toshiro Sugimoto; Keiji Isshiki; Masami Chin-Kanasaki; Masayoshi Sakaguchi; Naoto Kubota; Yasuo Terauchi; Takashi Kadowaki; Masakazu Haneda; Atsunori Kashiwagi; Daisuke Koya

Metabolic syndrome is associated with increased risk of chronic kidney disease, and the renal injury in patients with metabolic syndrome may be a result of altered renal lipid metabolism. We fed wild-type or insulin-sensitive heterozygous peroxisome proliferator-activated receptor gamma-deficient (PPARgamma(+/-)) mice a high-fat diet for 16 weeks. In wild-type mice, this diet induced core features of metabolic syndrome, subsequent renal lipid accumulation, and renal injury including glomerulosclerosis, interstitial fibrosis, and albuminuria. Renal lipogenesis accelerated, determined by increased renal mRNA expression of the lipogenic enzymes fatty acid synthase and acetyl-CoA carboxylase (ACC) and by increased ACC activity. In addition, renal lipolysis was suppressed, determined by reduced mRNA expression of the lipolytic enzyme carnitine palmitoyl acyl-CoA transferase 1 and by reduced activity of AMP-activated protein kinase. In PPARgamma(+/-) mice, renal injury, systemic metabolic abnormalities, renal accumulation of lipids, and the changes in renal lipid metabolism were attenuated. Thus, a high-fat diet leads to an altered balance between renal lipogenesis and lipolysis, subsequent renal accumulation of lipid, and renal injury. We suggest that renal lipid metabolism could serve as a new therapeutic target to prevent chronic kidney disease in patients with metabolic syndrome.


Diabetes | 2014

Linagliptin-Mediated DPP-4 Inhibition Ameliorates Kidney Fibrosis in Streptozotocin-Induced Diabetic Mice by Inhibiting Endothelial-to-Mesenchymal Transition in a Therapeutic Regimen

Keizo Kanasaki; Sen Shi; Megumi Kanasaki; Jianhua He; Takako Nagai; Yuka Nakamura; Yasuhito Ishigaki; Munehiro Kitada; Swayam Prakash Srivastava; Daisuke Koya

Kidney fibrosis is the final common pathway of all progressive chronic kidney diseases, of which diabetic nephropathy is the leading cause. Endothelial-to-mesenchymal transition (EndMT) has emerged as one of the most important origins of matrix-producing fibroblasts. Dipeptidyl peptidase-4 (DPP-4) inhibitors have been introduced into the market as antidiabetes drugs. Here, we found that the DPP-4 inhibitor linagliptin ameliorated kidney fibrosis in diabetic mice without altering the blood glucose levels associated with the inhibition of EndMT and the restoration of microRNA 29s. Streptozotocin-induced diabetic CD-1 mice exhibited kidney fibrosis and strong immunoreactivity for DPP-4 by 24 weeks after the onset of diabetes. At 20 weeks after the onset of diabetes, mice were treated with linagliptin for 4 weeks. Linagliptin-treated diabetic mice exhibited a suppression of DPP-4 activity/protein expression and an amelioration of kidney fibrosis associated with the inhibition of EndMT. The therapeutic effects of linagliptin on diabetic kidneys were associated with the suppression of profibrotic programs, as assessed by mRNA microarray analysis. We found that the induction of DPP-4 observed in diabetic kidneys may be associated with suppressed levels of microRNA 29s in diabetic mice; linagliptin restored microRNA 29s and suppressed DPP-4 protein levels. Using cultured endothelial cells, we found that linagliptin inhibited TGF-β2–induced EndMT, and such anti-EndMT effects of linagliptin were mediated through microRNA 29 induction. These results indicate the possible novel pleiotropic action of linagliptin to restore normal kidney function in diabetic patients with renal impairment.


Autophagy | 2012

Emerging role of autophagy in kidney function, diseases and aging

Tobias B. Huber; Charles L. Edelstein; Björn Hartleben; Ken Inoki; Man Jiang; Daisuke Koya; Shinji Kume; Wilfred Lieberthal; Nicolas Pallet; Alejandro Quiroga; Kameswaran Ravichandran; Katalin Susztak; Sei Yoshida; Zheng Dong

Autophagy is a highly conserved process that degrades cellular long-lived proteins and organelles. Accumulating evidence indicates that autophagy plays a critical role in kidney maintenance, diseases and aging. Ischemic, toxic, immunological, and oxidative insults can cause an induction of autophagy in renal epithelial cells modifying the course of various kidney diseases. This review summarizes recent insights on the role of autophagy in kidney physiology and diseases alluding to possible novel intervention strategies for treating specific kidney disorders by modifying autophagy.


Biochemical and Biophysical Research Communications | 2009

Exendin-4 has an anti-hypertensive effect in salt-sensitive mice model

Kunio Hirata; Shinji Kume; Shin-ichi Araki; Masayoshi Sakaguchi; Masami Chin-Kanasaki; Keiji Isshiki; Toshiro Sugimoto; Akira Nishiyama; Daisuke Koya; Masakazu Haneda; Atsunori Kashiwagi; Takashi Uzu

The improvement of salt-sensitive hypertension is a therapeutic target for various vascular diseases. Glucagon-like peptide 1 (GLP-1), an incretin peptide, has been reported to have natriuretic effect as well as blood glucose lowering effect, although its exact mechanism and clinical usefulness remain unclear. Here, we examined anti-hypertensive effect of exendin-4, a GLP-1 analog, in salt-sensitive obese db/db mice and angiotensin II (angII)-infused C57BLK6/J mice. The treatment of exendin-4 for 12 weeks inhibited the development of hypertension in db/db mice. In db/db mice, the urinary sodium excretion was delayed and blood pressure was elevated in response to a high-salt load, whereas these were attenuated by exendin-4. In db/db mice, intra-renal angII concentration was increased. Furthermore, exendin-4 prevented angII-induced hypertension in non-diabetic mice and inhibited angII-induced phosphorylation of ERK1/2 in cultured renal cells. Considered together, our results indicate that exendin-4 has anti-hypertensive effects through the attenuation of angII-induced high-salt sensitivity.


Diabetologia | 1991

Glucose enhances type IV collagen production in cultured rat glomerular mesangial cells

Masakazu Haneda; Ryuichi Kikkawa; Naoki Horide; Masaki Togawa; Daisuke Koya; Nobuyuki Kajiwara; A. Ooshima; Yukio Shigeta

SummaryType IV collagen production by cultured glomerular mesangial cells and the effect of glucose on it were evaluated in order to explore the possible contribution of mesangial cells to the accumulation of type IV collagen in mesangial matrix typically seen in diabetes. Type IV collagen was measured quantitatively by enzyme-linked immunosorbent assay. The majority of type IV collagen was secreted into culture media and secreted-type IV collagen increased with cell growth in early log phase and decreased in late log phase and after confluency. By exposing the cells to high concentrations of glucose (27.8 mmol/l), both secreted- and cell-associated-type IV collagens increased significantly compared with the cells cultured under normal glucose concentrations (5.6 mmol/l) or under equivalent concentrations of mannitol, resulting in a significant increase in total type IV collagen accumulation from 32.1±6.4 (under 5.6 mmol/l glucose) to 51.0±4.6 μg/dish (mean ± SD, n=4) on day 4, from 113.6±6.6 to 156.8±7.1 on day 6, from 248.5±15.2 to 310.0±12.6 on day 8 and from 372.4±14.8 to 507.9±17.2 on day 12. These results indicate the importance of glucose-induced alteration of mesangial cell function in the development of diabetic mesangial expansion.

Collaboration


Dive into the Daisuke Koya's collaboration.

Top Co-Authors

Avatar

Masakazu Haneda

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Atsunori Kashiwagi

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Keizo Kanasaki

Kanazawa Medical University

View shared research outputs
Top Co-Authors

Avatar

Takashi Uzu

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Shin-ichi Araki

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Shinji Kume

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Munehiro Kitada

Kanazawa Medical University

View shared research outputs
Top Co-Authors

Avatar

Toshiro Sugimoto

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Keiji Isshiki

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Ryuichi Kikkawa

Shiga University of Medical Science

View shared research outputs
Researchain Logo
Decentralizing Knowledge