Network


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

Hotspot


Dive into the research topics where Kanna Nagaishi is active.

Publication


Featured researches published by Kanna Nagaishi.


The Journal of Pathology | 2009

Enhancing epithelial engraftment of rat mesenchymal stem cells restores epithelial barrier integrity.

Takashi Yabana; Yoshiaki Arimura; Hiroki Tanaka; Akira Goto; Masayo Hosokawa; Kanna Nagaishi; Kentaro Yamashita; Hiroyuki Yamamoto; Yasushi Adachi; Yasushi Sasaki; Masaharu Isobe; Mineko Fujimiya; Kohzoh Imai; Yasuhisa Shinomura

The cellular origin, in vivo function and fate of donor bone marrow‐derived cells residing in the recipient intestinal epithelial cells, pericryptal myofibroblasts or endothelial cells remain obscure. Although ‘immunoprivileged’ mesenchymal stem cells (MSCs) are prime candidates for cell‐ and gene‐based therapy, their precise role in colitis remains largely undetermined. Using a dextran sulphate sodium (DSS) colitis with busulphan (BU)‐induced hypoplastic marrow model, we examined the therapeutic effects of MSC transplantation, focusing on the role of MSCs as both cell providers and immunomodulators. Donor‐derived MSCs were detected by eGFP immunofluorescence and fluorescence in situ hybridization for Y‐chromosome (Y‐FISH) analysis. Western blot analysis of apical‐most tight junction proteins was performed with antibodies against claudin‐2, ‐7, ‐8, ‐12, ‐13, ‐15 and ZO‐1. Cytokine and cell cycle profiles were analysed by semi‐quantitative RT‐PCR and flow cytometry. Susceptibility to DSS colitis was significantly increased by co‐existing BU‐induced bone marrow hypoplasia and this increase was significantly reduced by enhancing epithelial engraftment of MSCs, an effect depending on restoring epithelial barrier integrity rather than inhibiting host immune responses. We provide evidence that implicates MSCs in maintaining epithelial barrier function by reassembling apical‐most tight junction proteins, claudins. The therapeutic efficacy of extrinsic MSCs depends on enhancing epithelial engraftment in damaged crypts by busulphan conditioning. Such a role for the MSC‐derived intestinal cells in colitis therapy merits further examination and may offer a promising new treatment for inflammatory bowel disease (IBD). Copyright


PLOS ONE | 2013

Bone Marrow-Derived Microglia Infiltrate into the Paraventricular Nucleus of Chronic Psychological Stress-Loaded Mice

Koji Ataka; Akihiro Asakawa; Kanna Nagaishi; Kaori Kaimoto; Atsushi Sawada; Yuko Hayakawa; Ryota Tatezawa; Akio Inui; Mineko Fujimiya

Abstract Background Microglia of the central nervous system act as sentinels and rapidly react to infection or inflammation. The pathophysiological role of bone marrow-derived microglia is of particular interest because they affect neurodegenerative disorders and neuropathic pain. The hypothesis of the current study is that chronic psychological stress (chronic PS) induces the infiltration of bone marrow-derived microglia into hypothalamus by means of chemokine axes in brain and bone marrow. Methods and Findings Here we show that bone marrow-derived microglia specifically infiltrate the paraventricular nucleus (PVN) of mice that received chronic PS. Bone marrow derived-microglia are CX3CR1lowCCR2+CXCR4high, as distinct from CX3CR1highCCR2-CXCR4low resident microglia, and express higher levels of interleukin-1β (IL-1β) but lower levels of tumor necrosis factor-α (TNF-α). Chronic PS stimulates the expression of monocyte chemotactic protein-1 (MCP-1) in PVN neurons, reduces stromal cell-derived factor-1 (SDF-1) in the bone marrow and increases the frequency of CXCR4+ monocytes in peripheral circulation. And then a chemokine (C-C motif) receptor 2 (CCR2) or a β3-adrenoceptor blockade prevents infiltration of bone marrow-derived microglia in the PVN. Conclusion Chronic PS induces the infiltration of bone marrow-derived microglia into PVN, and it is conceivable that the MCP-1/CCR2 axis in PVN and the SDF-1/CXCR4 axis in bone marrow are involved in this mechanism.


Journal of Gastroenterology | 2014

Conditioned mesenchymal stem cells produce pleiotropic gut trophic factors.

Shuhei Watanabe; Yoshiaki Arimura; Kanna Nagaishi; Hiroyuki Isshiki; Kei Onodera; Masanao Nasuno; Kentaro Yamashita; Masashi Idogawa; Yasuyoshi Naishiro; Masaki Murata; Yasushi Adachi; Mineko Fujimiya; Kohzoh Imai; Yasuhisa Shinomura

BackgroundAlthough mounting evidence implicates mesenchymal stem cells (MSCs) in intestinal tissue repair, controversy remains regarding the engraftment, proliferation, and differentiation for repopulating MSCs in recipient tissues. Therefore, we investigated the paracrine and/or endocrine role of MSCs in experimental colitis.MethodsWe analyzed the therapeutic effects of MSC-conditioned medium (MSC-CM) on dextran sulfate sodium (DSS)- or 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis. We investigated the effects of MSC-CM on the epithelial cell viability, mobility, cell cycle, and cytokine production in ex vivo lamina propria/mesenteric lymphocytes, a macrophage cell line, and the mixed lymphocyte reaction. An optimal regimen against colitis was explored. The contents of MSC-CM were analyzed using a WNT signaling pathway polymerase chain reaction array, an inflammatory cytokines antibody array, and liquid chromatography-tandem mass spectrometry analysis.ResultsIndependent of the systemic administration route, MSC-CM concentrates were effective for the inductive phase of TNBS-induced colitis and for the recovery phase of DSS-induced colitis. Hypoxia appeared to be one of the optimal preconditioning factors assessed by cell motility and viability through activating the PI3K-Akt pathway in rat small intestine epithelial cells, IEC-6. Thus, Hypoxia had profound effects on the contents of MSC-CM, which comprised pleiotropic gut trophic factors involved in each wound healing process, including the anti-inflammatory, proliferative, and tissue remodeling phases.ConclusionsIdentification and optimization of potential gut trophic factors in MSC-CM is urgently needed to form the basis for new drug discovery and for optimizing cell-based therapies for inflammatory bowel disease.


Journal of Gastroenterology | 2015

Stem cell therapy for inflammatory bowel disease.

Kanna Nagaishi; Yoshiaki Arimura; Mineko Fujimiya

Inflammatory bowel disease (IBD) could be curable by “immune rest” and correction of the genetic predisposition inherent in allogeneic hematopoietic stem cell transplantation. However, balancing risks against benefits remains challenging. The application of mesenchymal stem cells (MSCs) serving as a site-regulated “drugstore” is a recent concept, which suggests the possibility of an alternative treatment for many intractable diseases such as IBD. Depending on the required function of MSC, such as a cell provider, immune moderator, and/or trophic resource, MSC therapy should be optimized to maximize its therapeutic benefit. Therapeutic effects do not always require full engraftment of MSCs. Therefore, optimization of pleiotropic gut trophic factors produced by MSCs, which favoring not only regulating immune responses but also promoting tissue repair, must directly enhance new drug discoveries for treatment of IBD. Stem cell biology holds great promise for a new era of cell-based therapy, sparking considerable interest among scientists, clinicians, and patients. However, the translational arm of stem cell science remains in a relatively primitive state. Although several clinical studies using MSCs have been initiated, early results suggest several inherent problems. In each study, optimization of MSC therapy appears to be the most urgent problem, and can be resolved only by scientifically unveiling the mechanisms of therapeutic action. In the present review, the authors outline how such information would facilitate the critical steps in the paradigm shift from basic research on stem cell biology to clinical practice of regenerative medicine for conquering IBD in the near future.


Pain | 2014

Suppression of bone marrow–derived microglia in the amygdala improves anxiety-like behavior induced by chronic partial sciatic nerve ligation in mice

Atsushi Sawada; Yukitoshi Niiyama; Koji Ataka; Kanna Nagaishi; Michiaki Yamakage; Mineko Fujimiya

Summary Bone marrow–derived microglia aggregated in the central nucleus of the amygdala are involved in the chronic neuropathic pain–induced anxiety. ABSTRACT Chronic neuropathic pain causes abnormal sensitivities such as hyperalgesia and allodynia, and emotional abnormalities such as anxiety and depression. Although spinal cord microglia are involved in abnormal sensitivity to neuropathic pain, no previous studies have examined the mechanism of neuropathic pain–induced anxiety. Here, we examined the involvement of bone marrow (BM)–derived microglia aggregated in the amygdalae of mice with chronic neuropathic pain in the development of anxiety‐like behavior. We prepared partial sciatic nerve ligations (PSNL) in mice that received bone marrow transplantation from green fluorescent protein (GFP)–Tg mice after irradiation with head protection, and examined GFP‐positive microglia in the central nuclei of the amygdalae (CeA). On day 28 after PSNL, BM‐derived microglia aggregated in the CeA concurrent with anxiety‐like behavior. BM‐derived microglia in the CeA highly expressed interleukin (IL)–1&bgr; and C‐C chemokine receptor type 2 (CCR2). In addition, neurons in the CeA highly expressed monocyte chemotactic protein–1 (MCP‐1), a ligand for CCR2, in PSNL‐treated mice compared to sham‐operated mice, suggesting that the MCP‐1/CCR2 axis is involved in the recruitment of BM‐derived microglia. Oral administration of a CCR2 antagonist decreased the number of BM‐derived microglia in the CeA, and successfully reversed the anxiety‐like behavior and hypersensitivity to mechanical stimuli in PSNL‐treated mice. Microinjections of an IL‐1&bgr; receptor antagonist directly into the CeA successfully reversed the anxiety‐like behavior in the PSNL‐treated mice even though the neuropathic pain persisted. These results suggest that the recruitment of BM‐derived microglia to the CeA via the MCP‐1/CCR2 axis and neuron–microglia interactions might be important in the pathogenesis of neuropathic pain–induced anxiety.


Journal of Gastroenterology | 2011

Myogenic lineage differentiated mesenchymal stem cells enhance recovery from dextran sulfate sodium-induced colitis in the rat

Hiroki Tanaka; Yoshiaki Arimura; Takashi Yabana; Akira Goto; Masayo Hosokawa; Kanna Nagaishi; Kentaro Yamashita; Hiroyuki Yamamoto; Yasushi Sasaki; Mineko Fujimiya; Kohzoh Imai; Yasuhisa Shinomura

BackgroundAlthough mounting evidence implicates mesenchymal stem cells (MSCs) in intestinal tissue repair, uncertainty remains concerning the distribution, function, and fate of repopulating MSCs in recipient colonic tissues. Therefore, we investigated the role of transplanted MSCs in the repair phase of DSS colitis.MethodsLacZ-labeled rat MSCs were transplanted into rats with colitis induced by 4% DSS on day 2. Regular water replaced the DSS solution on day 6. Therapeutic effect was evaluated on day 9 by clinicopathologic and growth factor/cytokine expression profiles. We analyzed the Notch signaling pathway by Western blotting and characterized immunofluorescence of lacZ-labeled MSCs with confocal laser microscopy. In vivo differentiation of MSC was confirmed by transmission electron microscopy (TEM).ResultsRecovery of colitis was modestly but significantly promoted by MSC transplantation due to proceeding cell cycle and inhibiting apoptosis in the epithelia. Tgfa mRNA expression increased significantly, while Notch signaling was inhibited in the colonic tissues with MSC transplantation. β-Galactosidase-positive cells, which expressed α-SMA, desmin, and vimentin, were infrequently detected in the lamina propria stroma. DSS exposure in vitro proved to be the most potent inducer for α-SMA in MSCs where TEM demonstrated myogenic lineage differentiation.ConclusionsWe found that MSCs transplantation modestly promoted the repair of DSS colitis. The donor-derived MSCs were likely reprogrammed to differentiate to myogenic lineage cells by cues from the micro milieu. Further characterization of these cells is warranted as a basis for applying cell-based therapy for inflammatory bowel disease.


Scientific Reports | 2016

Bone marrow-derived mesenchymal stem cells improve diabetes-induced cognitive impairment by exosome transfer into damaged neurons and astrocytes

Masako Nakano; Kanna Nagaishi; Naoto Konari; Yuki Saito; Takako Chikenji; Yuka Mizue; Mineko Fujimiya

The incidence of dementia is higher in diabetic patients, but no effective treatment has been developed. This study showed that rat bone marrow mesenchymal stem cells (BM-MSCs) can improve the cognitive impairments of STZ-diabetic mice by repairing damaged neurons and astrocytes. The Morris water maze test demonstrated that cognitive impairments induced by diabetes were significantly improved by intravenous injection of BM-MSCs. In the CA1 region of the hippocampus, degeneration of neurons and astrocytes, as well as synaptic loss, were prominent in diabetes, and BM-MSC treatment successfully normalized them. Since a limited number of donor BM-MSCs was observed in the brain parenchyma, we hypothesized that humoral factors, especially exosomes released from BM-MSCs, act on damaged neurons and astrocytes. To investigate the effectiveness of exosomes for treatment of diabetes-induced cognitive impairment, exosomes were purified from the culture media and injected intracerebroventricularly into diabetic mice. Recovery of cognitive impairment and histological abnormalities similar to that seen with BM-MSC injection was found following exosome treatment. Use of fluorescence-labeled exosomes demonstrated that injected exosomes were internalized into astrocytes and neurons; these subsequently reversed the dysfunction. The present results indicate that exosomes derived from BM-MSCs might be a promising therapeutic tool for diabetes-induced cognitive impairment.


Scientific Reports | 2016

Mesenchymal stem cell therapy ameliorates diabetic nephropathy via the paracrine effect of renal trophic factors including exosomes

Kanna Nagaishi; Yuka Mizue; Takako Chikenji; Miho Otani; Masako Nakano; Naoto Konari; Mineko Fujimiya

Bone marrow-derived mesenchymal stem cells (MSCs) have contributed to the improvement of diabetic nephropathy (DN); however, the actual mediator of this effect and its role has not been characterized thoroughly. We investigated the effects of MSC therapy on DN, focusing on the paracrine effect of renal trophic factors, including exosomes secreted by MSCs. MSCs and MSC-conditioned medium (MSC-CM) as renal trophic factors were administered in parallel to high-fat diet (HFD)-induced type 2 diabetic mice and streptozotocin (STZ)-induced insulin-deficient diabetic mice. Both therapies showed approximately equivalent curative effects, as each inhibited the exacerbation of albuminuria. They also suppressed the excessive infiltration of BMDCs into the kidney by regulating the expression of the adhesion molecule ICAM-1. Proinflammatory cytokine expression (e.g., TNF-α) and fibrosis in tubular interstitium were inhibited. TGF-β1 expression was down-regulated and tight junction protein expression (e.g., ZO-1) was maintained, which sequentially suppressed the epithelial-to-mesenchymal transition of tubular epithelial cells (TECs). Exosomes purified from MSC-CM exerted an anti-apoptotic effect and protected tight junction structure in TECs. The increase of glomerular mesangium substrate was inhibited in HFD-diabetic mice. MSC therapy is a promising tool to prevent DN via the paracrine effect of renal trophic factors including exosomes due to its multifactorial action.


Hepatology | 2014

Mesenchymal stem cell therapy ameliorates diabetic hepatocyte damage in mice by inhibiting infiltration of bone marrow–derived cells

Kanna Nagaishi; Koji Ataka; Eijiro Echizen; Yoshiaki Arimura; Mineko Fujimiya

Although mesenchymal stem cells (MSCs) have been implicated in hepatic injury, the mechanism through which they contribute to diabetic liver disease has not been clarified. In this study, we investigated the effects of MSC therapy on diabetic liver damage with a focus on the role of bone‐marrow–derived cells (BMDCs), which infiltrate the liver, and elucidated the mechanism mediating this process. Rat bone‐marrow (BM)‐derived MSCs were administered to high‐fat diet (HFD)‐induced type 2 diabetic mice and streptozotocin (STZ)‐induced insulin‐deficient diabetic mice. MSC‐conditioned medium (MSC‐CM) was also administered to examine the trophic effects of MSCs on liver damage. Therapeutic effects of MSCs were analyzed by assessing serum liver enzyme levels and histological findings. Kinetic and molecular profiles of BMDCs in the liver were evaluated using BM‐chimeric mice. Curative effects of MSC and MSC‐CM therapies were similar because both ameliorated the aggravation of aspartate aminotransferase and alanine aminotransferase at 8 weeks of treatment, despite persistent hyperlipidemia and hyperinsulinemia in HFD‐diabetic mice and persistent hyperglycemia in STZ‐diabetic mice. Furthermore, both therapies suppressed the abnormal infiltration of BMDCs into the liver, reversed excessive expression of proinflammatory cytokines in parenchymal cells, and regulated proliferation and survival signaling in the liver in both HFD‐ and STZ‐diabetic mice. In addition to inducing hepatocyte regeneration in STZ‐diabetic mice, both therapies also prevented excessive lipid accumulation and apoptosis of hepatocytes and reversed insulin resistance (IR) in HFD‐diabetic mice. Conclusion: MSC therapy is a powerful tool for repairing diabetic hepatocyte damage by inhibiting inflammatory reactions induced by BMDCs and IR. These effects are likely the result of humoral factors derived from MSCs. (Hepatology 2014;59:1816–1829)


Stem Cells | 2014

Mesenchymal Stem Cells Cancel Azoxymethane‐Induced Tumor Initiation

Masanao Nasuno; Yoshiaki Arimura; Kanna Nagaishi; Hiroyuki Isshiki; Kei Onodera; Suguru Nakagaki; Shuhei Watanabe; Masashi Idogawa; Kentaro Yamashita; Yasuyoshi Naishiro; Yasushi Adachi; Hiromu Suzuki; Mineko Fujimiya; Kohzoh Imai; Yasuhisa Shinomura

The role of mesenchymal stem cells (MSCs) in tumorigenesis remains controversial. Therefore, our goal was to determine whether exogenous MSCs possess intrinsic antineoplastic or proneoplastic properties in azoxymethane (AOM)‐induced carcinogenesis. Three in vivo models were studied: an AOM/dextran sulfate sodium colitis‐associated carcinoma model, an aberrant crypt foci model, and a model to assess the acute apoptotic response of a genotoxic carcinogen (AARGC). We also performed in vitro coculture experiments. As a result, we found that MSCs partially canceled AOM‐induced tumor initiation but not tumor promotion. Moreover, MSCs inhibited the AARGC in colonic epithelial cells because of the removal of O6‐methylguanine (O6MeG) adducts through O6MeG‐DNA methyltransferase activation. Furthermore, MSCs broadly affected the cell‐cycle machinery, potentially leading to G1 arrest in vivo. Coculture of IEC‐6 rat intestinal cells with MSCs not only arrested the cell cycle at the G1 phase, but also induced apoptosis. The anti‐carcinogenetic properties of MSCs in vitro required transforming growth factor (TGF)‐β signaling because such properties were completely abrogated by absorption of TGF‐β under indirect coculture conditions. MSCs inhibited AOM‐induced tumor initiation by preventing the initiating cells from sustaining DNA insults and subsequently inducing G1 arrest in the initiated cells that escaped from the AARGC. Furthermore, tumor initiation perturbed by MSCs might potentially dysregulate WNT and TGF‐β‐Smad signaling pathways in subsequent tumorigenesis. Obtaining a better understanding of MSC functions in colon carcinogenesis is essential before commencing the broader clinical application of promising MSC‐based therapies for cancer‐prone patients with inflammatory bowel disease. Stem Cells 2014;32:913–925

Collaboration


Dive into the Kanna Nagaishi's collaboration.

Top Co-Authors

Avatar

Mineko Fujimiya

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar

Yoshiaki Arimura

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kentaro Yamashita

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Koji Ataka

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hiroyuki Isshiki

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar

Masako Nakano

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar

Takako Chikenji

Sapporo Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge