Network


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

Hotspot


Dive into the research topics where Miwako Katagi is active.

Publication


Featured researches published by Miwako Katagi.


American Journal of Physiology-endocrinology and Metabolism | 2011

Inactivation of TNF-α ameliorates diabetic neuropathy in mice

Isamu Yamakawa; Hideto Kojima; Tomoya Terashima; Miwako Katagi; Jiro Oi; Hiroshi Urabe; Mitsuru Sanada; Hiromichi Kawai; Lawrence Chan; Hitoshi Yasuda; Hiroshi Maegawa; Hiroshi Kimura

Tumor necrosis factor (TNF)-α is a potent proinflammatory cytokine involved in the pathogenesis of diabetic neuropathy. We inactivated TNF-α to determine if it is a valid therapeutic target for the treatment of diabetic neuropathy. We effected the inactivation in diabetic neuropathy using two approaches: by genetic inactivation of TNF-α (TNF-α(-/-) mice) or by neutralization of TNF-α protein using the monoclonal antibody infliximab. We induced diabetes using streptozotocin in wild-type and TNF-α(-/-) mice. We measured serum TNF-α concentration and the level of TNF-α mRNA in the dorsal root ganglion (DRG) and evaluated nerve function by a combination of motor (MNCV) and sensory (SNCV) nerve conduction velocities and tail flick test, as well as cytological analysis of intraepidermal nerve fiber density (IENFD) and immunostaining of DRG for NF-κB p65 serine-276 phosphorylated and cleaved caspase-3. Compared with nondiabetic mice, TNF-α(+/+) diabetic mice displayed significant impairments of MNCV, SNCV, tail flick test, and IENFD as well as increased expression of NF-κB p65 and cleaved caspase-3 in their DRG. In contrast, although nondiabetic TNF-α(-/-) mice showed mild abnormalities of IENFD under basal conditions, diabetic TNF-α(-/-) mice showed no evidence of abnormal nerve function tests compared with nondiabetic mice. A single injection of infliximab in diabetic TNF-α(+/+) mice led to suppression of the increased serum TNF-α and amelioration of the electrophysiological and biochemical deficits for at least 4 wk. Moreover, the increased TNF-α mRNA expression in diabetic DRG was also attenuated by infliximab, suggesting infliximabs effects may involve the local suppression of TNF-α. Infliximab, an agent currently in clinical use, is effective in targeting TNF-α action and expression and amelioration of diabetic neuropathy in mice.


Bone | 2010

Malfunction of Bone Marrow Derived Osteoclasts and the Delay of Bone Fracture Healing in Diabetic Mice

Toshiyuki Kasahara; Sinji Imai; Hideto Kojima; Miwako Katagi; Hiroshi Kimura; Lawrence Chan; Yoshitaka Matsusue

It is well known that bone fracture healing is delayed in diabetes mellitus, but the mechanism remains to be elucidated. Since several studies have demonstrated that diabetes causes abnormalities in bone marrow-derived cells, we used the streptozotocin (STZ)-induced diabetic mouse model after bone marrow transfer from green fluorescent protein (GFP) transgenic mice, and examined fracture healing. Compared with nondiabetic mice, diabetic mice at 3 weeks after fracture showed a decrease in mineralized callus, with the remainder consisting of cartilage. Bone formation parameters and mineralization rate were not altered in the STZ mice, but bone resorption parameters were significantly decreased. Therefore, the delayed bone formation in the STZ mice may have resulted from an impairment of cartilage resorption. Interestingly, we found that 80% of the osteoclasts in the callus were derived from bone marrow and the sizes of the osteoclasts as well as the resorption pits formed were significantly smaller in the diabetic mice. Moreover, transcript analysis using RNA isolated by laser capture microdissection (LCM) showed that the expression of DC-STAMP, a putative pivotal gene for osteoclast fusion, was decreased in osteoclasts from diabetic mice. Since the sustainability of osteoclast function depends on the controlled renewal of multinuclear osteoclasts, impaired osteoclast function in diabetes may contribute to decreased cartilage resorption and delayed endochondral ossification.


PLOS ONE | 2012

Brain-derived neurotrophic factor from bone marrow-derived cells promotes post-injury repair of peripheral nerve.

Yoshinori Takemura; Shinji Imai; Hideto Kojima; Miwako Katagi; Isamu Yamakawa; Toshiyuki Kasahara; Hiroshi Urabe; Tomoya Terashima; Hitoshi Yasuda; Lawrence Chan; Hiroshi Kimura; Yoshitaka Matsusue

Brain-derived neurotrophic factor (BDNF) stimulates peripheral nerve regeneration. However, the origin of BNDF and its precise effect on nerve repair have not been clarified. In this study, we examined the role of BDNF from bone marrow-derived cells (BMDCs) in post-injury nerve repair. Control and heterozygote BDNF knockout mice (BDNF+/−) received a left sciatic nerve crush using a cerebral blood clip. Especially, for the evaluation of BDNF from BMDCs, studies with bone marrow transplantation (BMT) were performed before the injury. We evaluated nerve function using a rotarod test, sciatic function index (SFI), and motor nerve conduction velocity (MNCV) simultaneously with histological nerve analyses by immunohistochemistry before and after the nerve injury until 8 weeks. BDNF production was examined by immunohistochemistry and mRNA analyses. After the nerve crush, the controls showed severe nerve dysfunction evaluated at 1 week. However, nerve function was gradually restored and reached normal levels by 8 weeks. By immunohistochemistry, BDNF expression was very faint before injury, but was dramatically increased after injury at 1 week in the distal segment from the crush site. BDNF expression was mainly co-localized with CD45 in BMDCs, which was further confirmed by the appearance of GFP-positive cells in the BMT study. Variant analysis of BDNF mRNA also confirmed this finding. BDNF+/− mice showed a loss of function with delayed histological recovery and BDNF+/+→BDNF+/− BMT mice showed complete recovery both functionally and histologically. These results suggested that the attenuated recovery of the BDNF+/− mice was rescued by the transplantation of BMCs and that BDNF from BMDCs has an essential role in nerve repair.


Nature Communications | 2013

Haematopoietic cells produce BDNF and regulate appetite upon migration to the hypothalamus

Hiroshi Urabe; Hideto Kojima; Lawrence Chan; Tomoya Terashima; Nobuhiro Ogawa; Miwako Katagi; Kazunori Fujino; Asako Kumagai; Hiromichi Kawai; Akihiro Asakawa; Akio Inui; Hitoshi Yasuda; Yutaka Eguchi; Kazuhiro Oka; Hiroshi Maegawa; Atsunori Kashiwagi; Hiroshi Kimura

Brain-derived neurotrophic factor (BDNF) suppresses food intake by acting on neurons in the hypothalamus. Here we show that BDNF-producing haematopoietic cells control appetite and energy balance by migrating to the hypothalamic paraventricular nucleus. These haematopoietic-derived paraventricular nucleus cells produce microglial markers and make direct contacts with neurons in response to feeding status. Mice with congenital BDNF deficiency, specifically in haematopoietic cells, develop hyperphagia, obesity and insulin resistance. These abnormalities are ameliorated by bone marrow transplantation with wild-type bone marrow cells. Furthermore, when injected into the third ventricle, wild-type bone marrow mononuclear cells home to the paraventricular nucleus and reverse the hyperphagia of BDNF-deficient mice. Our results suggest a novel mechanism of feeding control based on the production of BDNF by haematopoietic cells and highlight a potential new therapeutic route for the treatment of obesity.


FEBS Letters | 2014

Hyperglycemia induces abnormal gene expression in hematopoietic stem cells and their progeny in diabetic neuropathy

Miwako Katagi; Tomoya Terashima; Junko Okano; Hiroshi Urabe; Yuki Nakae; Nobuhiro Ogawa; Jun Udagawa; Hiroshi Maegawa; Kazuhiro Matsumura; Lawrence Chan; Hideto Kojima

Diabetic peripheral neuropathy is a major chronic diabetic complication. We have previously shown that in type 1 diabetic streptozotocin‐treated mice, insulin‐ and TNF‐α co‐expressing bone marrow‐derived cells (BMDCs) induced by hyperglycemia travel to nerve tissues where they fuse with nerve cells, causing premature apoptosis and nerve dysfunction. Here we show that similar BMDCs also occur in type 2 diabetic high‐fat diet (HFD) mice. Furthermore, we found that hyperglycemia induces the co‐expression of insulin and TNF‐α in c‐kit+Sca‐1+lineage− (KSL) progenitor cells, which maintain the same expression pattern in the progeny, which in turn participates in the fusion with neurons when transferred to normoglycemic animals.


Journal of Gastroenterology and Hepatology | 2006

Peptide YY3‐36 and pancreatic polypeptide suppress food intake

Akihiro Asakawa; Masaharu Uemoto; Naohiko Ueno; Miwako Katagi; Mineko Fujimiya; Kazunori Fujino; Noriko Kodama; Hiroaki Nanba; Ruka Sakamaki; Naotaka Shinfuku; Michael M. Meguid; Akio Inui

To the Editor , Here, we show that peptide YY (PYY) 3–36 and pancreatic polypeptide (PP) suppress food intake in both lean mice and ob/ob obese mice. Pancreatic polypeptide and PYY are 36 amino acid peptides that belong to a family of peptides including neuropeptide Y (NPY). Since the discovery of leptin and ghrelin, much progress has been made in the study of controlling energy homeostasis. Until now, various peptides, including NPY, ghrelin, agoutirelated protein (AGRP), orexin, melanin-concentrating hormone (MCH), leptin, α -melanocyte-stimulating hormone (MSH), cocaineand amphetamine-regulated transcript (CART), cholecystokinin (CCK), corticotropin-releasing factor (CRF) and urocortin, have been shown to be involved in the regulation of food intake. Tschop et al . reported that gut hormone PYY3-36 does not suppress food intake in rodents and they cannot replicate the anorexigenic effects of PYY3-36 reported by Batterham et al . 1–3


Journal of Gastroenterology and Hepatology | 2011

Homing of the bone marrow-derived interstitial cells of Cajal is decreased in diabetic mouse intestine.

Yimin Li; Hideto Kojima; Kazunori Fujino; Kazuhiro Matsumura; Miwako Katagi; Hiroshi Urabe; Lawrence Chan; Yutaka Eguchi; Linghui Zhao; Hiroshi Kimura

Background:  Interstitial cells of Cajal (ICCs), which express c‐Kit receptor tyrosine kinase (KIT), play an important role in gastrointestinal motility. Loss of ICCs likely contributes to diabetic gastrointestinal motility disorder, however, the mechanism of attrition remains unknown. Here, we test the hypothesis that the bone marrow‐derived progenitors are an important source of intestinal ICCs and that decreased homing of these progenitors in diabetes contributes to ICC diminution.


PLOS ONE | 2016

Hyperglycemia Induces Skin Barrier Dysfunctions with Impairment of Epidermal Integrity in Non-Wounded Skin of Type 1 Diabetic Mice.

Junko Okano; Hideto Kojima; Miwako Katagi; Takahiko Nakagawa; Yuki Nakae; Tomoya Terashima; Takeshi Kurakane; Mamoru Kubota; Hiroshi Maegawa; Jun Udagawa

Diabetes causes skin complications, including xerosis and foot ulcers. Ulcers complicated by infections exacerbate skin conditions, and in severe cases, limb/toe amputations are required to prevent the development of sepsis. Here, we hypothesize that hyperglycemia induces skin barrier dysfunction with alterations of epidermal integrity. The effects of hyperglycemia on the epidermis were examined in streptozotocin-induced diabetic mice with/without insulin therapy. The results showed that dye leakages were prominent, and transepidermal water loss after tape stripping was exacerbated in diabetic mice. These data indicate that hyperglycemia impaired skin barrier functions. Additionally, the distribution of the protein associated with the tight junction structure, tight junction protein-1 (ZO-1), was characterized by diffuse and significantly wider expression in the diabetic mice compared to that in the control mice. In turn, epidermal cell number was significantly reduced and basal cells were irregularly aligned with ultrastructural alterations in diabetic mice. In contrast, the number of corneocytes, namely, denucleated and terminally differentiated keratinocytes significantly increased, while their sensitivity to mechanical stress was enhanced in the diabetic mice. We found that cell proliferation was significantly decreased, while apoptotic cells were comparable in the skin of diabetic mice, compared to those in the control mice. In the epidermis, Keratin 5 and keratin 14 expressions were reduced, while keratin 10 and loricrin were ectopically induced in diabetic mice. These data suggest that hyperglycemia altered keratinocyte proliferation/differentiation. Finally, these phenotypes observed in diabetic mice were mitigated by insulin treatment. Reduction in basal cell number and perturbation of the proliferation/differentiation process could be the underlying mechanisms for impaired skin barrier functions in diabetic mice.


Biochemical and Biophysical Research Communications | 2015

Epidermis–dermis junction as a novel location for bone marrow-derived cells to reside in response to ionizing radiation

Junko Okano; Hideto Kojima; Miwako Katagi; Yuki Nakae; Tomoya Terashima; Takahiko Nakagawa; Takeshi Kurakane; Naoki Okamoto; Keita Morohashi; Hiroshi Maegawa; Jun Udagawa

Bone marrow-derived cells (BMDCs) can migrate into the various organs in the mice irradiated by ionizing radiation (IR). However, it may not be the case in the skin. While IR is used for bone marrow (BM) transplantation, studying with the epidermal sheets demonstrated that the BMDC recruitment is extraordinarily rare in epidermis in the mouse. Herein, using the chimera mice with BM from green fluorescent protein (GFP) transgenic mice, we simply examined if BMDCs migrate into any layers in the total skin, as opposed to the epidermal sheets, in response to IR. Interestingly, we identified the presence of GFP-positive (GFP(+)) cells in the epidermis-dermis junction in the total skin sections although the epidermal cell sheets failed to have any GFP cells. To examine a possibility that the cells in the junction could be mechanically dissociated during separating epidermal sheets, we then salvaged such dissociated cells and examined its characteristics. Surprisingly, some GFP(+) cells were found in the salvaged cells, indicating that these cells could be derived from BM. In addition, such BMDCs were also associated with inflammation in the junction. In conclusion, BMDCs can migrate to and reside in the epidermis-dermis junction after IR.


Molecular therapy. Nucleic acids | 2018

Gene Therapy for Neuropathic Pain through siRNA-IRF5 Gene Delivery with Homing Peptides to Microglia

Tomoya Terashima; Nobuhiro Ogawa; Yuki Nakae; Toshiyuki Sato; Miwako Katagi; Junko Okano; Hiroshi Maegawa; Hideto Kojima

Astrocyte- and microglia-targeting peptides were identified and isolated using phage display technology. A series of procedures, including three cycles of both in vivo and in vitro biopanning, was performed separately in astrocytes and in M1 or M2 microglia, yielding 50–58 phage plaques in each cell type. Analyses of the sequences of this collection identified one candidate homing peptide targeting astrocytes (AS1[C-LNSSQPS-C]) and two candidate homing peptides targeting microglia (MG1[C-HHSSSAR-C] and MG2[C-NTGSPYE-C]). To determine peptide specificity for the target cell in vitro, each peptide was synthesized and introduced into the primary cultures of astrocytes or microglia. Those peptides could bind to the target cells and be selectively taken up by the corresponding cell, namely, astrocytes, M1 microglia, or M2 microglia. To confirm cell-specific gene delivery to M1 microglia, the complexes between peptide MG1 and siRNA-interferon regulatory factor 5 were prepared and intrathecally injected into a mouse model of neuropathic pain. The complexes successfully suppressed hyperalgesia with high efficiency in this neuropathic pain model. Here, we describe a novel gene therapy for the treatment neuropathic pain, which has a high potential to be of clinical relevance. This strategy will ensure the targeted delivery of therapeutic genes while minimizing side effects to non-target tissues or cells.

Collaboration


Dive into the Miwako Katagi's collaboration.

Top Co-Authors

Avatar

Hideto Kojima

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Tomoya Terashima

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Hiroshi Maegawa

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Yuki Nakae

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Lawrence Chan

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hiroshi Kimura

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Hiroshi Urabe

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Junko Okano

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hitoshi Yasuda

Shiga University of Medical Science

View shared research outputs
Researchain Logo
Decentralizing Knowledge