Network


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

Hotspot


Dive into the research topics where Kyosuke Yamanishi is active.

Publication


Featured researches published by Kyosuke Yamanishi.


Comprehensive Psychiatry | 2014

The impacts of elevated autism spectrum disorder traits on clinical and psychosocial features and long-term treatment outcome in adult patients with obsessive-compulsive disorder.

Hironori Mito; Naomi Matsuura; Keiitiro Mukai; Yoshinobu Yanagisawa; Akihiro Nakajima; Mikuni Motoyama; Ayako Arikawa; Kyosuke Yamanishi; Hisato Matsunaga

BACKGROUND While a close relation between obsessive-compulsive disorder (OCD) and autism spectrum disorder (ASD) has been pointed out, there are few studies that have investigated whether highly elevated ASD traits may have significant impacts on clinical and psychosocial features as well as long-term treatment outcome in adult OCD patients. METHODS We assessed ASD traits using the Autism Spectrum Quotient (AQ) in 81 Japanese patients with OCD. The relation between degree of ASD traits and clinical and psychosocial variables and the 48-week treatment outcomes was analyzed in the subjects. RESULTS A substantial proportion of the subjects showed higher ASD traits (35%) with more severe depressive or pervasive anxiety status, and social impairments and lower QOL compared to other OCD individuals. However, elevated ASD traits may exert rather smaller impact on the OCD phenomenology along with on the long-term treatment outcome than expected. CONCLUSIONS Elevated ASD traits may further emphasize the general psychopathological and socio-dysfunctional features rather than clinical aspects associated with OCD. Co-existing depressive or anxious symptom severity may further exacerbate the core-deficits related to ASD pathology. Thus the assessment of ASD traits should be important for understanding the clinical and psychosocial features and treatment responses in OCD patients.


Clinical Cancer Research | 2016

Augmentation of Immune Checkpoint Cancer Immunotherapy with IL18

Zhifeng Ma; Wen Li; Shinichi Yoshiya; Yunfeng Xu; Masaki Hata; Yosif El-Darawish; Tzvetanka Markova; Kyosuke Yamanishi; Hiromichi Yamanishi; Hideaki Tahara; Yoshimasa Tanaka; Haruki Okamura

Purpose: Recent clinical trials and animal models demonstrated that immune checkpoint blockade enhanced effector cell responses and tumor rejection; however, further development and improvement of cancer immunotherapy is necessary for more favorable objective responses. In this study, we examined the effect of IL18 on the antitumor effect of immune checkpoint inhibitors. Experimental Design: We examined the effect of IL18 on the peritoneal dissemination of CT-26 cells or tail vein injection metastasis of B16/F10 cells using antiprogrammed death-1 ligand-1 (αPD-L1) and/or anti-CTL–associated antigen-4 (αCTLA-4) mAbs. Result: Massive ascites developed after intraperitoneal inoculation of CT-26, resulting in animal death within 30 days. Treatment of mice with αPD-L1 and/or αCTLA-4 significantly prolonged their survival, and a combination of the antibodies and IL18 provided a much greater therapeutic benefit. The combination modality led to the accumulation of precursor of mature natural killer (pre-mNK) cells in the peritoneal cavity together with increased CD8+ T and decreased CD4+CD25+Foxp3+ T cells. Depletion of the pre-mNK cells abrogated the therapeutic effects and increased the number of CD4+CD25+Foxp3+ T cells. The combination treatment also suppressed tail vein injection metastasis of B16/F10 cells. Conclusions: The results demonstrated that IL18 enhanced therapeutic effects of immune checkpoint blockade against peritoneal dissemination of carcinoma or tail vein injection metastasis of melanoma through accumulation of pre-mNK cells, memory-type CD8+ T cells, and suppression of CD4+CD25+Foxp3+ T cells. A combination of immune checkpoint inhibitors with IL18 may give a suggestion to the development of next-generation cancer immunotherapy. Clin Cancer Res; 22(12); 2969–80. ©2016 AACR.


International Journal of Molecular Medicine | 2013

Genetic analysis of genes causing hypertension and stroke in spontaneously hypertensive rats

Hideyuki Yamamoto; Daisuke Okuzaki; Kyosuke Yamanishi; Yunfeng Xu; Yuko Watanabe; M. Yoshida; Akifumi Yamashita; Naohisa Goto; Seiji Nishiguchi; Kazunori Shimada; Hiroshi Nojima; Teruo Yasunaga; Haruki Okamura; Hisato Matsunaga; Hiromichi Yamanishi

Spontaneously hypertensive rats (SHR) and stroke-prone SHR (SHRSP) are frequently used as model rats not only in studies of essential hypertension and stroke, but also in studies of attention deficit hyperactivity disorder (ADHD). Normotensive Wistar-Kyoto rats (WKY) are normally used as controls in these studies. In this study, using these rats, we aimed to identify the genes causing hypertension and stroke, as well as the genes involved in ADHD. Since adrenal gland products can directly influence cardiovascular, endocrine and sympathetic nervous system functions, gene expression profiles in the adrenal glands of the 3 rat strains were examined using genome-wide microarray technology when the rats were 3 and 6 weeks of age, a period in which the rats are considered to be in a pre-hypertensive state. Gene expression profiles were compared between SHR and WKY and between SHRSP and SHR. A total of 353 genes showing more than a 4-fold increase or less than a 4-fold decrease in expression were isolated and candidate genes were selected as significantly enriched genes. SHR-specific genes isolated when the rats were 3 weeks of age contained 12 enriched genes related to transcriptional regulatory activity and those isolated when the rats were 6 weeks of age contained 6 enriched genes related to the regulation of blood pressure. SHRSP-specific genes isolated when the rats were 3 weeks of age contained 4 enriched genes related to the regulation of blood pressure and those isolated when the rats were 6 weeks of age contained 4 enriched genes related to the response to steroid hormone stimulus. Ingenuity pathway analysis of enriched SHR-specific genes revealed that 2 transcriptional regulators, cAMP responsive element modulator (Crem) and Fos-like antigen 1 (Fosl1), interact with blood pressure-regulating genes, such as neurotensin (Nts), apelin (Apln) and epoxide hydrolase 2, cytoplasmic (Ephx2). Similar analyses of SHRSP-specific genes revealed that angiotensinogen (Agt), one of the blood pressure-regulating genes, plays pivotal roles among SHRSP-specific genes. Moreover, genes associated with ADHD, such as low density lipoprotein receptor (Ldlr) and Crem, are discussed.


Journal of Hypertension | 2014

Chymase inhibition improves vascular dysfunction and survival in stroke-prone spontaneously hypertensive rats

Shinji Takai; Denan Jin; Hong Chen; Wen Li; Hideyuki Yamamoto; Kyosuke Yamanishi; Mizuo Miyazaki; Hideaki Higashino; Hiromichi Yamanishi; Haruki Okamura

Objective: To clarify the role of chymase in hypertension, we evaluated the effect of a chymase inhibitor, TY-51469, on vascular dysfunction and survival in stroke-prone spontaneously hypertensive rats (SHR-SP). Methods: SHR-SP were treated with TY-51469 (1 mg/kg per day) or placebo from 4 to 12 weeks old or until death. Wistar–Kyoto rats were used as a normal group. Results: SBP was significantly higher in both the placebo and TY-51469 groups than in the normal group, but there was no significant difference between the two treatment groups. Plasma renin, angiotensin-converting enzyme activity and angiotensin II levels were not different between the placebo and TY-51469 groups. In contrast, vascular chymase-like activity was significantly higher in the placebo than in the normal group, but it was reduced by TY-51469. Acetylcholine-induced vascular relaxation was significantly higher in the TY-51469 group than in the placebo group. There was significant augmentation of the number of monocytes/macrophages and matrix metalloproteinase-9 activity in aortic tissue from the placebo group compared with the normal group, and these changes were attenuated by TY-51469. There were also significant increases in mRNA levels of monocyte chemoattractant protein-1 and tumor necrosis factor-&agr; in the placebo group that were attenuated by TY-51469. Cumulative survival was significantly prolonged in the TY-51469 group compared with the placebo group. Conclusion: Chymase might play an important role in vascular dysfunction via augmentation both of matrix metalloproteinase-9 activity and monocyte/macrophage accumulation in SHR-SP, and its inhibition may be useful for preventing vascular remodeling and prolonging survival.


American Journal of Physiology-heart and Circulatory Physiology | 2016

Dysfunction of mitochondria and deformed gap junctions in the heart of IL-18-deficient mice

Wen Li; Denan Jin; Masaki Hata; Shinji Takai; Kyosuke Yamanishi; Weili Shen; Yosif El-Darawish; Hiromichi Yamanishi; Haruki Okamura

Interleukin-18 (IL-18) was discovered as an interferon-γ-inducing factor and has been regarded as a proinflammatory cytokine. However, IL-18 is ubiquitously expressed both in immune/inflammatory cells and in nonimmune cells, and its biological roles have not been sufficiently elucidated. Here, we demonstrate that IL-18-deficient [IL-18 knockout (KO)] mice have heart abnormalities that may be related to impaired autophagy. In endurance running tests, IL-18KO mice ran significantly shorter distances compared with wild-type (WT) mice. Echocardiographs indicated disability in the systolic and diastolic functions of the IL-18KO mouse heart. Immunostaining of connexin 43 showed heterogeneous localization of gap junctions in the lateral membranes of the IL-18KO cardiac myocytes. Western blotting analysis revealed decreased phosphorylated connexin 43 in the IL-18KO heart. Electron microscopy revealed unusual localization of intercalated disks, swollen or damaged mitochondria, and broad, indistinct Z-lines in the IL-18KO heart. In accordance with the morphological observation, mitochondrial respiratory function, including that of complexes I and IV, was impaired, and production of reactive oxygen species was augmented in IL-18KO hearts. Notably, levels of LC3-II were markedly lower in the IL-18KO hearts than in WT hearts. In the culture of cardiac myocytes of IL-18KO neonates, exogenous IL-18 upregulated LC3-II and increased the number of intact mitochondria with high mitochondrial membrane potential. These results indicated that IL-18 has roles apart from those as a proinflammatory cytokine in cardiac myocytes and suggested that IL-18 contributes to the homeostatic maintenance of mitochondrial function and gap-junction turnover in cardiac myocytes, possibly by upregulating autophagy.


PLOS ONE | 2013

Regulation of Development of CD56brightCD11c+ NK-like Cells with Helper Function by IL-18

Wen Li; Akico Okuda; Hideyuki Yamamoto; Kyosuke Yamanishi; Nobuyuki Terada; Hiromichi Yamanishi; Yoshimasa Tanaka; Haruki Okamura

Human γδ T cells augment host defense against tumors and infections, and might have a therapeutic potential in immunotherapy. However, mechanism of γδ T cell proliferation is unclear, and therefore it is difficult to prepare sufficient numbers of γδ T cells for clinical immunotherapy. Recently, natural killer (NK)-like CD56brightCD11c+ cells were shown to promote the proliferation of γδ T cells in an IL-18-dependent manner. In this study, we demonstrated that the NK-like CD56brightCD11c+ cells could directly interact with γδ T cells to promote their sustained expansion, while conventional dendritic cells (DCs), IFN-α-induced DCs, plasmacytoid DCs or monocytes did not. We also examined the cellular mechanism underlying the regulation of CD56brightCD11c+ cells. CD14+ monocytes pre-incubated with IL-2/IL-18 formed intensive interactions with CD56intCD11c+ cells to promote their differentiation to CD56brightCD11c+ cells with helper function. The development of CD56brightCD11c+ cells was suppressed in an IFN-α dependent manner. These results indicate that CD14+ monocytes pretreated with IL-2/IL-18, but neither DCs nor monocytes, play a determining role on the development and proliferation of CD56brightCD11c+ cells, which in turn modulate the expansion of γδ T cells. CD56brightCD11c+ NK-like cells may be a novel target for immunotherapy utilizing γδ T cells, by overcoming the limitation of γδ T cells proliferation.


International Journal of Molecular Medicine | 2015

Genetic analysis of genes causing hypertension and stroke in spontaneously hypertensive rats: Gene expression profiles in the kidneys

Yuko Watanabe; M. Yoshida; Kyosuke Yamanishi; Hideyuki Yamamoto; Daisuke Okuzaki; Hiroshi Nojima; Teruo Yasunaga; Haruki Okamura; Hisato Matsunaga; Hiromichi Yamanishi

Spontaneously hypertensive rats (SHRs) and stroke-prone SHRs (SHRSP) are frequently used as models not only of essential hypertension and stroke, but also of attention-deficit hyperactivity disorder (ADHD). Normotensive Wistar-Kyoto (WKY) rats are normally used as controls in these studies. In the present study, we aimed to identify the genes causing hypertension and stroke, as well as the genes involved in ADHD using these rats. We previously analyzed gene expression profiles in the adrenal glands and brain. Since the kidneys can directly influence the functions of the cardiovascular, endocrine and sympathetic nervous systems, gene expression profiles in the kidneys of the 3 rat strains were examined using genome-wide microarray technology when the rats were 3 and 6 weeks old, a period in which rats are considered to be in a pre-hypertensive state. Gene expression profiles were compared between the SHRs and WKY rats and also between the SHRSP and SHRs. A total of 232 unique genes showing more than a 4-fold increase or less than a 4-fold decrease in expression were isolated as SHR- and SHRSP-specific genes. Candidate genes were then selected using two different web tools: the 1st tool was the Database for Annotation, Visualization and Integrated Discovery (DAVID), which was used to search for significantly enriched genes and categorized them using Gene Ontology (GO) terms, and the 2nd was Ingenuity Pathway Analysis (IPA), which was used to search for interactions among SHR- and also SHRSP-specific genes. The analyses of SHR-specific genes using IPA revealed that B-cell CLL/lymphoma 6 (Bcl6) and SRY (sex determining region Y)-box 2 (Sox2) were possible candidate genes responsible for causing hypertension in SHRs. Similar analyses of SHRSP-specific genes revealed that angiotensinogen (Agt), angiotensin II receptor-associated protein (Agtrap) and apolipoprotein H (Apoh) were possible candidate genes responsible for triggering strokes. Since our results revealed that SHRSP-specific genes isolated from the kidneys of rats at 6 weeks of age, included 6 genes related to Huntingtons disease, we discussed the genetic association between ADHD and Huntingtons disease.


Journal of Leukocyte Biology | 2018

Frontline Science: IL‐18 primes murine NK cells for proliferation by promoting protein synthesis, survival, and autophagy

Yosif El-Darawish; Wen Li; Kyosuke Yamanishi; Magdalena Pencheva; Naoto Oka; Hiromichi Yamanishi; Tomohiro Matsuyama; Yoshimasa Tanaka; Nagahiro Minato; Haruki Okamura

Combined stimulation by IL‐2 and IL‐18 effectively promotes proliferation of NK cells, whereas singular stimulation does not. In this study, synergistic effects of these cytokines on NK cells proliferation was analyzed, focusing on the roles of IL‐18. In splenic resting NK cells from IL‐18KO mice, IL‐18 rapidly activated NF‐κB independently of IL‐2, and activated or up‐regulated various molecules downstream of PI3K/AKT and mTOR, including S6, Bcl‐XL, ATG5, and LC3II, accompanying increases in cell growth and survival. Thus, IL‐18 alone was revealed to augment various cellular processes (gene transcription, protein synthesis, survival) in the absence or presence of IL‐2. Notably, combined IL‐18 and IL‐2 promoted autophagosome formation. In addition, priming NK cells with IL‐18 augmented IL‐2R, especially CD25, and enabled cells to respond to IL‐2, resulting in activation of STAT3 and STAT5, followed by increase of cyclin B1 leading to proliferation. However, IL‐2 alone failed to activate STAT3 or STAT5 in resting IL18KO NK cells. These results clarify the distinct roles of IL‐2 and IL‐18 in NK cell proliferation, and the intrinsic roles of IL‐18 in various cellular processes, suggesting a range of functions of IL‐18 expressed in an array of nonhematopoietic cells.


Neuroscience Letters | 2017

Interleukin-18 and its receptor are expressed in gonadotropin-releasing hormone neurons of mouse and rat forebrain

Sachi Kuwahara-Otani; Seishi Maeda; Kimiko Kobayashi; Yusuke Minato; Koichi Tanaka; Kyosuke Yamanishi; Masaki Hata; Wen Li; Tetsu Hayakawa; Koichi Noguchi; Haruki Okamura; Hideshi Yagi

Interleukin-18 (IL-18) is a pro-inflammatory cytokine and an important mediator of peripheral inflammation and host immune response. IL-18 functions through its binding with the IL-18 receptor (IL-18R), which consists of two chains, an IL-18-binding α chain (IL-18Rα) and a signaling β chain. IL-18 and IL-18R are expressed in the brain; however, limited information is available on IL-18R expression and the role of IL-18 in neurosecretory cells. In the present study, we used immunohistochemical techniques to investigate the distribution of IL-18Rα and IL-18 in the hypothalamus of male mice and rats. IL-18Rα-positive and IL-18-positive perikarya and fibers were found scattered throughout the medial septal nucleus, the nuclei of the vertical and horizontal limbs of the diagonal band, the organum vasculosum of the laminae terminalis, the preoptic area, and the anterior hypothalamic area. It is well known that gonadotropin-releasing hormone (GnRH) neuronal somata and/or fibers are found in these regions. Therefore, we performed double-label immunofluorescence for IL-18Rα/IL-18 and GnRH. IL-18Rα was expressed in approximately 60% of GnRH-immunopositive perikarya, and IL-18 was distributed in all GnRH-immunopositive perikarya. These observations suggest that IL-18 exerts direct effects upon the GnRH neuron via IL-18Rα and acts on GnRH neurons through an autocrine or paracrine pathway.


Journal of Chemical Neuroanatomy | 2016

Fine structure of interleukin 18 (IL-18) receptor-immunoreactive neurons in the retrosplenial cortex and its changes in IL18 knockout mice.

Tetsu Hayakawa; Masaki Hata; Sachi Kuwahara-Otani; Kyosuke Yamanishi; Hideshi Yagi; Haruki Okamura

Interleukin 18 (IL-18) participates in the inflammatory immune response of lymphocytes. Delay in learning or memory are common in the IL-18 knockout mouse. Many IL-18-immunoreactive neurons are found in the retrosplenial cortex (RSC) and the subiculum. These neurons also contain the IL-18 receptor. We determined the location and the ultrastructure of the IL-18 receptor-immunoreactive neurons in the RSC and observed changes in the IL-18 receptor-immunoreactive neurons of the IL-18 knockout mouse. The IL-18 receptor-immunoreactive neurons were found specifically in layer V of the granular RSC. They were medium-sized neurons with a light oval nucleus and had little cytoplasm with many free ribosomes, rough endoplasmic reticulum and many mitochondria, but no Nissl bodies. The number of axosomatic terminals was about six per section. The IL-18 receptor-immunoreactive neurons were not found in the RSC in the IL-18 knockout mouse at 5 or 9 weeks of age. However, many small electron-dense neurons were found in layer V. Both the nucleus and cytoplasm were electron-dense, but not necrotic. The mitochondria and rough endoplasmic reticulum were swollen. The IL-18 receptor-immunoreactive neurons were presumed to be degenerating. The degeneration of the IL18-receptor-immunoreactive neurons in the RSC may cause the abnormal behaviors of the IL-18 knockout mice.

Collaboration


Dive into the Kyosuke Yamanishi's collaboration.

Top Co-Authors

Avatar

Haruki Okamura

Hyogo College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wen Li

Hyogo College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yuko Watanabe

Jichi 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

Tetsu Hayakawa

Hyogo College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge