Network


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

Hotspot


Dive into the research topics where Hitoshi Harada is active.

Publication


Featured researches published by Hitoshi Harada.


Journal of Cell Science | 2006

Phosphorylation of paracellin-1 at Ser217 by protein kinase A is essential for localization in tight junctions.

Akira Ikari; Satomi Matsumoto; Hitoshi Harada; Kuniaki Takagi; Hisayoshi Hayashi; Yuichi Suzuki; Masakuni Degawa; Masao Miwa

Although paracellin-1 (PCLN-1) is known to have a crucial role in the control of Mg2+ reabsorption in the kidney, the molecular pathways involved in the regulation of PCLN-1 have not been clarified. We used FLAG-tagged PCLN-1 to investigate these pathways further, and found that PCLN-1 is phosphorylated at Ser217 by protein kinase A (PKA) under physiological conditions in Madin-Darby canine kidney (MDCK) cells. PCLN-1 expression decreased Na+ permeability, resulting in a decrease in the transepithelial electrical resistance (TER). By contrast, PCLN-1 enhanced transepithelial Mg2+ transport. PKA inhibitors, N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide dihydrochloride (H-89) and myristoylated protein kinase A inhibitor 14-22 amide PKI, and an adenylate cyclase inhibitor, 2′,5′-dideoxy adenosine (DDA), reduced the phosphoserine level of PCLN-1. The inhibitory effect of DDA was rescued by 8-bromoadenosine-3′,5′-cyclic monophosphate (8-Br-cAMP). PKA and adenylate cyclase inhibitors decreased transepithelial Mg2+ transport and TER. Dephosphorylated PCLN-1 moved from detergent-insoluble to soluble fractions and was dissociated from ZO-1. A fusion protein of PCLN-1 with glutathione-S-transferase revealed that Ser217 was phosphorylated by PKA. Phosphorylated PCLN-1 was localized in the tight junction (TJ) along with ZO-1, whereas dephosphorylated PCLN-1 and the S217A mutant were translocated into the lysosome. The degradation of dephosphorylated PCLN-1 and S217A mutant was inhibited by chloroquine, a specific lysosome inhibitor. Thus, the PKA-dependent phosphorylation of Ser217 in PCLN-1 is essential for its localization in the TJ and transepithelial Mg2+ transport.


Biochimica et Biophysica Acta | 2010

γ-Irradiation induces P2X7 receptor-dependent ATP release from B16 melanoma cells

Yasuhiro Ohshima; Mitsutoshi Tsukimoto; Takato Takenouchi; Hitoshi Harada; Akina Suzuki; Mitsuru Sato; Hiroshi Kitani; Shuji Kojima

BACKGROUND Ionizing irradiation causes not only growth arrest and cell death, but also release of growth factors or signal transmitters, which promote cancer malignancy. Extracellular ATP controls cancer growth through activation of purinoceptors. However, there is no report of radiation-induced ATP release from cancer cells. Here, we examined gamma-irradiation-induced ATP release and its mechanism in B16 melanoma. METHODS Extracellular ATP was measured by luciferin-luciferase assay. To investigate mechanism of radiation-induced ATP release, we pharmacologically inhibited the ATP release and established stable P2X(7) receptor-knockdown B16 melanoma cells using two short hairpin RNAs targeting P2X(7) receptor. RESULTS Cells were exposed to 0.5-8 Gy of gamma-rays. Extracellular ATP was increased, peaking at 5 min after 0.5 Gy irradiation. A selective P2X(7) receptor channel antagonist, but not anion transporter inhibitors, blocked the release of ATP. Further, radiation-induced ATP release was significantly decreased in P2X(7) receptor-knockdown cells. Our results indicate that gamma-irradiation evokes ATP release from melanoma cells, and P2X(7) receptor channel plays a significant role in mediating the ATP release. GENERAL SIGNIFICANCE We suggest that extracellular ATP could be a novel intercellular signaling molecule released from cancer cells when cells are exposed to ionizing radiation.


Journal of Biological Chemistry | 2010

Involvement of SLC17A9-dependent vesicular exocytosis in the mechanism of ATP release during T cell activation.

Akihiro Tokunaga; Mitsutoshi Tsukimoto; Hitoshi Harada; Yoshinori Moriyama; Shuji Kojima

Recent reports have shown that T cell receptor (TCR)-dependent ATP release from T cells is involved in production of interleukin-2 (IL-2) through activation of P2 receptors. Stimulation of TCR induces ATP release from T cells through gap junction hemichannels and maxianion channels, at least in part. However, the mechanisms of ATP release from activated T cells are not fully understood. Here, we studied the mechanisms of ATP release during TCR-dependent T cell activation by investigating the effects of various inhibitors on TCR-dependent ATP release from murine T cells. We found that not only anion channel and gap junction hemichannel inhibitors, but also exocytosis inhibitors suppressed the ATP release. These results suggest that ATP release from murine T cells is regulated by various mechanisms, including exocytosis. An inhibitor of exocytosis, bafilomycin A, significantly blocked TCR signaling, such as Ca2+ elevation and IL-2 production. Furthermore, bafilomycin A, ectonucleotidase, and P2Y6 receptor antagonist significantly inhibited production of pro-inflammatory cytokines from external antigen-restimulated splenocytes, indicating that vesicular exocytosis-mediated purinergic signaling has a significant role in TCR-dependent cytokine production. We also detected vesicular ATP in murine T cells and human T lymphoma Jurkat cells, both of which also expressed mRNA of SLC17A9, a vesicular nucleotide transporter. Knockdown of SLC17A9 in Jurkat cells markedly reduced ATP release and cytosolic Ca2+ elevation after TCR stimulation, suggesting involvement of SLC17A9-dependent vesicular exocytosis in ATP release and T cell activation. In conclusion, vesicular exocytosis of ATP appears to play a role in T cell activation and immune responses.


PLOS ONE | 2013

Autocrine Regulation of Macrophage Activation via Exocytosis of ATP and Activation of P2Y11 Receptor

Hayato Sakaki; Mitsutoshi Tsukimoto; Hitoshi Harada; Yoshinori Moriyama; Shuji Kojima

It is important to understand the mechanisms that regulate macrophage activation to establish novel therapies for inflammatory diseases, such as sepsis; a systemic inflammatory response syndrome generally caused by bacterial lipopolysaccharide (LPS). In this study, we investigated the involvement of extracellular ATP-mediated autocrine signaling in LPS-induced macrophage activation. We show here that ATP release via exocytosis, followed by activation of P2Y11 receptor, is a major pathway of the macrophage activation, leading to release of cytokines. Treatment of human monocyte THP-1 cells with LPS induced rapid ATP release from cells, and this release was blocked by knockdown of SLC17A9 (vesicular nucleotide transporter, VNUT), which is responsible for exocytosis of ATP. ATP-enriched vesicles were found in cytosol of THP-1 cells. These data suggest the involvement of vesicular exocytosis in the release of ATP. Knockdown of SLC17A9, the P2Y11 antagonist NF157 or knockdown of P2Y11 receptor significantly suppressed both M1-type polarization and IL-6 production in THP-1 cells, indicating an important role of activation of P2Y11 receptor by released ATP in macrophage activation. Next, the effect of NF157 on LPS-induced immune activation was examined in vivo. Administration of LPS to mice caused increase of serum IL-1ß, IL-6, IL-12 and TNF-alpha levels at 3–24 h after the administration. Pre-treatment of LPS-treated mice with NF157 suppressed both elevation of proinflammatory cytokines in serum and M1 polarization of peritoneal/spleen macrophages. Moreover, post-treatment with NF157 at 30 min after administration of LPS also suppressed the elevation of serum cytokines levels. We conclude that vesicular exocytosis of ATP and autocrine, positive feedback through P2Y11 receptors is required for the effective activation of macrophages. Consequently, P2Y11 receptor antagonists may be drug candidates for treatment of inflammatory diseases such as sepsis.


Biochemical and Biophysical Research Communications | 2012

Regulation of P2X7-dependent inflammatory functions by P2X4 receptor in mouse macrophages.

Ayumi Kawano; Mitsutoshi Tsukimoto; Daisuke Mori; Taisei Noguchi; Hitoshi Harada; Takato Takenouchi; Hiroshi Kitani; Shuji Kojima

Activation of the P2X7 receptor of macrophages plays an important role in inflammation. We recently reported that co-expression of P2X4 receptor with P2X7 receptor facilitates P2X7 receptor-mediated cell death via Ca(2+) influx. However, it remained unclear whether P2X4 receptor is involved in P2X7 receptor-mediated inflammatory responses, such as cytokine production. Here, we present evidence that P2X4 receptor modulates P2X7 receptor-dependent inflammatory functions. Treatment of mouse macrophage RAW264.7 cells with 1mM ATP induced high mobility group box 1 (HMGB1) release and IL-1β production via activation of P2X7 receptor. Knockdown of P2X4 receptor or removal of extracellular Ca(2+) suppressed ATP-induced release of both HMGB1 and IL-1β. On the other hand, knockdown of P2X4 receptor or removal of extracellular Ca(2+) enhanced P2X7-dependent LC3-II expression (an index of autophagy), suggesting that P2X4 receptor suppresses P2X7-mediated autophagy. Since LC3-II expression was inhibited by pretreatment with antioxidant and NADPH oxidase inhibitor, we examined P2X7-mediated production of reactive oxygen species (ROS). We found that activation of P2X7 receptor-mediated production of ROS was significantly facilitated in P2X4-knockdown cells, suggesting that co-expression of P2X4 receptor with P2X7 receptor may suppress anti-inflammatory function-related autophagy via suppression of ROS production. We conclude that co-expression of P2X4 receptor with P2X7 receptor enhances P2X7-mediated inflammation through both facilitation of release of cytokines and suppression of autophagy.


Journal of Cell Science | 2012

Autocrine regulation of TGF-β1-induced cell migration by exocytosis of ATP and activation of P2 receptors in human lung cancer cells.

Erina Takai; Mitsutoshi Tsukimoto; Hitoshi Harada; Keisuke Sawada; Yoshinori Moriyama; Shuji Kojima

Summary TGF-&bgr;1 plays a key role in cancer progression through induction of various biological effects, including cell migration. Extracellular nucleotides, such as ATP, released from cells play a role in signaling through activation of P2 receptors. We show here that exocytosis of ATP followed by activation of P2 receptors play a key role in TGF-&bgr;1-induced actin remodeling associated with cell migration. Treatment with TGF-&bgr;1 facilitated migration of human lung cancer A549 cells, which was blocked by pretreatment with ecto-nucleotidase and P2 receptor antagonists. ATP and P2 agonists facilitated cell migration. TGF-&bgr;1-induced actin remodeling, which contributes to cell migration, was also suppressed by pretreatment with ecto-nucleotidase and P2 receptor antagonists. Knockdown of P2X7 receptor suppressed TGF-&bgr;1-induced migration and actin remodeling. These results indicate the involvement of TGF-&bgr;1-induced ATP release in cell migration, at least in part, through activation of P2X7 receptors. TGF-&bgr;1 caused release of ATP from A549 cells within 10 minutes. Both ATP-enriched vesicles and expression of a vesicular nucleotide transporter (VNUT) SLC17A9, which is responsible for exocytosis of ATP, were found in cytosol of A549 cells. TGF-&bgr;1 failed to induce release of ATP from SLC17A9-knockdown cells. TGF-&bgr;1-induced cell migration and actin remodeling were also decreased in SLC17A9-knockdown cells. These results suggest the importance of exocytosis of ATP in cell migration. We conclude that autocrine signaling through exocytosis of ATP and activation of P2 receptors is required for the amplification of TGF-&bgr;1-induced migration of lung cancer cells.


Biochemical and Biophysical Research Communications | 2012

Involvement of P2X4 receptor in P2X7 receptor-dependent cell death of mouse macrophages.

Ayumi Kawano; Mitsutoshi Tsukimoto; Taisei Noguchi; Noriyuki Hotta; Hitoshi Harada; Takato Takenouchi; Hiroshi Kitani; Shuji Kojima

Interaction of P2X7 receptor with P2X4 receptor has recently been suggested, but it remains unclear whether P2X4 receptor is involved in P2X7 receptor-mediated events, such as cell death of macrophages induced by high concentrations of extracellular ATP. Here, we present evidence that P2X4 receptor does play a role in P2X7 receptor-dependent cell death. Treatment of mouse macrophage RAW264.7 cells with 1mM ATP induced Ca(2+) influx, non-selective large pore formation, activation of extracellular signal-regulated protein kinase (ERK) 1/2 and p38 mitogen-activated protein kinase (MAPK), and cell death via activation of P2X7 receptor. P2X4-knockdown cells, established by transfecting RAW264.7 cells with two short hairpin RNAs (shRNAs) targeting P2X4 receptor, showed a decrease of the initial peak of intracellular Ca(2+) after treatment with ATP, though pore formation and the P2X7-mediated activation of ERK1/2 and p38 MAPK were not affected. Intriguingly, P2X4 knockdown resulted in significant suppression of cell death induced by ATP or P2X7 agonist BzATP. In conclusion, our results suggest that P2X4 receptor is involved in P2X7 receptor-mediated cell death, but not pore formation or MAPK signaling.


Biochemical and Biophysical Research Communications | 2013

P2X4 receptor regulates P2X7 receptor-dependent IL-1β and IL-18 release in mouse bone marrow-derived dendritic cells

Hayato Sakaki; Takuya Fujiwaki; Mitsutoshi Tsukimoto; Ayumi Kawano; Hitoshi Harada; Shuji Kojima

Activation of P2X7 receptor of dendritic cells plays a significant role in inflammation through production of cytokines such as IL-1β, and recent studies have suggested structural and functional interactions of P2X7 receptor with P2X4 receptor in macrophages. However, it is unknown whether P2X4 receptor modulates P2X7 functions in dendritic cells. Here, we present evidence that expression of P2X4 receptor is required for P2X7 receptor-dependent IL-1β and IL-18 release in mouse bone marrow-derived dendritic cells (BMDCs). We confirmed expression of both P2X7 receptor and P2X4 receptor in BMDCs. Treatment of BMDCs with 3 mM ATP caused a transient, P2X4-dependent elevation, or spike, of intracellular Ca(2+) level [Ca(2+)]i, followed by the sustained P2X7-dependent increase of [Ca(2+)]i. We performed knockdown of P2X4 receptor in BMDCs by transfection with short hairpin RNA targeting this receptor. The ATP-induced initial peak of [Ca(2+)]i was decreased in P2X4-knockdown cells (P2X4-KD). Further, we found that ATP-induced IL-1β and IL-18 release from LPS-primed BMDCs was suppressed by pretreatment with P2X7 antagonist A438079 or P2X4 antagonist TNP-ATP. The P2X7-dependent IL-1β and IL-18 release was significantly lower in P2X4-KD cells. Chelation of intracellular Ca(2+) also caused suppression of ATP-induced IL-1β and IL-18 release. These results suggest that P2X4 receptor-induced Ca(2+) influx is required for effective production of IL-1β and IL-18 via activation of P2X7 receptor in BMDCs. We conclude that co-expression of P2X4 receptor with P2X7 receptor in dendritic cells leads to enhancement of inflammation through facilitation of P2X7-dependent release of pro-inflammatory cytokines.


Purinergic Signalling | 2014

Autocrine signaling via release of ATP and activation of P2X7 receptor influences motile activity of human lung cancer cells

Erina Takai; Mitsutoshi Tsukimoto; Hitoshi Harada; Shuji Kojima

Extracellular nucleotides, such as ATP, are released from cells and play roles in various physiological and pathological processes through activation of P2 receptors. Here, we show that autocrine signaling through release of ATP and activation of P2X7 receptor influences migration of human lung cancer cells. Release of ATP was induced by stimulation with TGF-β1, which is a potent inducer of cell migration, in human lung cancer H292 cells, but not in noncancerous BEAS-2B cells. Treatment of H292 cells with a specific antagonist of P2X7 receptor resulted in suppression of TGF-β1-induced migration. PC-9 human lung cancer cells released a large amount of ATP under standard cell culture conditions, and P2X7 receptor-dependent dye uptake was observed even in the absence of exogenous ligand, suggesting constitutive activation of P2X7 receptor in this cell line. PC-9 cells showed high motile activity, which was inhibited by treatment with ecto-nucleotidase and P2X7 receptor antagonists, whereas a P2X7 receptor agonist enhanced migration. PC-9 cells also harbor a constitutively active mutation in epidermal growth factor receptor (EGFR). Treatment with EGFR tyrosine kinase inhibitor AG1478 suppressed both cell migration and P2X7 receptor expression in PC-9 cells. Compared to control PC-9 cells, cells treated with P2X7 antagonist exhibited broadened lamellipodia around the cell periphery, while AG1478-treated cells lacked lamellipodia. These results indicate that P2X7-mediated signaling and EGFR signaling may regulate migration of PC-9 cells through distinct mechanisms. We propose that autocrine ATP-P2X7 signaling is involved in migration of human lung cancer cells through regulation of actin cytoskeleton rearrangement.


Biochemical and Biophysical Research Communications | 2009

Blockade of murine T cell activation by antagonists of P2Y6 and P2X7 receptors

Mitsutoshi Tsukimoto; Akihiro Tokunaga; Hitoshi Harada; Shuji Kojima

Extracellular nucleotides and their metabolites activate ionotropic P2X and metabotropic P2Y receptors on the surface of various types of cells. Here, we investigated the involvement of P2X and P2Y receptor-mediated signaling in TCR-dependent T cell activation. Murine T cells were activated by stimulation of TCR, and both CD25 expression and interleukin (IL)-2 production were observed in activated T cells. Ecto-nucleotidase apyrase and P2Y6 antagonist MRS2578 significantly blocked the increases of both CD25 expression and IL-2 production, and P2X7 antagonists A438079 and oxidized ATP inhibited IL-2 production rather than CD25 expression, suggesting the involvement of P2Y6 and P2X7 receptors in different processes of T cell activation. MRS2578 also blocked TCR-dependent elevation of cytosolic Ca2+ in T cells. The P2X7 and P2Y6 receptors were expressed in murine CD4 T cells. In conclusion, our results indicate that activation of P2Y6 and P2X7 receptors contributes to T cell activation via TCR.

Collaboration


Dive into the Hitoshi Harada's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shuji Kojima

Tokyo University of Science

View shared research outputs
Top Co-Authors

Avatar

Akira Ikari

Gifu Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Takato Takenouchi

National Agriculture and Food Research Organization

View shared research outputs
Top Co-Authors

Avatar

Erina Takai

Tokyo University of Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hiroshi Kitani

National Agriculture and Food Research Organization

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ayumi Kawano

Tokyo University of Science

View shared research outputs
Researchain Logo
Decentralizing Knowledge