Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Asian J Endosc Surg ; 17(1): e13274, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38212269

ABSTRACT

BACKGROUND: Pelvic lymph node dissection is a procedure performed in gastroenterological surgery, urology, and gynecology. However, due to discrepancies in the understanding of pelvic anatomy among these departments, cross-disciplinary discussions have not been easy. Recently, with the rapid spread of robotic surgery, the importance of visual information in understanding pelvic anatomy has become even more significant. In this project, we attempted to clarify a shared understanding of pelvic anatomy through cross-disciplinary discussions. METHOD: From May 2020 to November 2021, a total of 11 discussions were held entirely online with 5 colorectal surgery specialists, 4 urologists, and 4 gynecologists. The discussions focused on evidence from each specialty and surgical videos, aiming to create a universally understandable pelvic anatomical illustration. RESULTS: The common area of dissection recognized across the three departments was identified as the obturator lymph nodes. A dynamic illustration of pelvic anatomy was created. In addition to a bird's-eye view of the pelvis, a pelvic half view was developed to enhance understanding of the deeper pelvic anatomy. The following insights were incorporated into the illustration: (1) the cardinal ligament in gynecology partly overlaps with the vesicohypogastric fascia in colorectal surgery; (2) the obturator lymph nodes continue cephalad into the fossa of Marcille in urology; and (3) the deep uterine vein in gynecology corresponds to the inferior vesical vein in colorectal surgery. CONCLUSION: Based on the dynamic illustration of pelvic anatomy from cross-disciplinary discussions, we anticipate advancements in pelvic lymph node dissection aiming for curative and safe outcomes.


Subject(s)
Colorectal Surgery , Gynecology , Robotics , Urology , Humans , Anatomy, Regional , Lymph Node Excision/methods , Lymph Nodes/pathology , Reference Standards
2.
PLoS One ; 16(6): e0253646, 2021.
Article in English | MEDLINE | ID: mdl-34166439

ABSTRACT

PURPOSE: Since December 2019, coronavirus disease 2019 (COVID-19) has spread rapidly across the world. During the pandemic, physicians in our hospital have had to respond both to the issue of treating the patients and the increasing domestic burden associated with social disruption. The purpose of this study was to assess how much the burden on our doctors, especially female doctors, was increasing. MATERIAL AND METHODS: The Physicians' Career Support Committee in Sapporo Medical University conducted a questionnaire survey. The questionnaire inquired about a wide range of subjects with regard to working style and family life during the first and second waves of the COVID-19 pandemic, and was sent to all medical/dental physicians working in Sapporo Medical University. RESULTS: A total of 266 (42.7%) physicians in our hospital responded to our questionnaire and the data for 264 data were analyzed. The total numbers of males, females, and others, including those who did not want to specify, were 178 (67.4%), 82 (31.0%), and 4 (1.5%), respectively. Among them, 62 (23.5%) and 23 (8.7%) answered that their domestic burden was slightly or markedly increased. The increase in the domestic burden showed a significant difference between genders (p = 0.04). Even after correction for background differences using multivariate analysis, being female (p<0.001), having child dependents (p<0.001), and treating COVID-19 patients (p = 0.03) were significantly related to an increased domestic burden. Regarding family style, 58.1% of the physician-fathers were from two-income families (i.e., families with both parents in employment), and they answered that their partner mainly cared for the children. In contrast, 97.3% of physician-mothers were from two-income families, and 94.6% of the physician-mothers had to take care of children by themselves. CONCLUSION: Physician-mothers are caught in a dilemma between an increased home burden and clinical duties in the hospital, with a significantly higher ratio than physician-fathers during the pandemic. As we showed, female doctors could have not continued their careers and take responsible positions in the same way as male doctors. This is a social risk in the timing of a crisis, such as a pandemic.


Subject(s)
COVID-19 , Mothers , Pandemics , Physicians, Women , SARS-CoV-2 , Surveys and Questionnaires , Women, Working , Adult , Aged , Female , Humans , Japan/epidemiology , Middle Aged
4.
Cell Mol Gastroenterol Hepatol ; 9(2): 277-293, 2020.
Article in English | MEDLINE | ID: mdl-31622786

ABSTRACT

BACKGROUND & AIMS: Ral guanosine triphosphatase-activating protein α2 (RalGAPα2) is the major catalytic subunit of the negative regulators of the small guanosine triphosphatase Ral, a member of the Ras subfamily. Ral regulates tumorigenesis and invasion/metastasis of some cancers; however, the role of Ral in colitis-associated cancer (CAC) has not been investigated. We aimed to elucidate the role of Ral in the mechanism of CAC. METHODS: We used wild-type (WT) mice and RalGAPα2 knockout (KO) mice that showed Ral activation, and bone marrow chimeric mice were generated as follows: WT to WT, WT to RalGAPα2 KO, RalGAPα2 KO to WT, and RalGAPα2 KO to RalGAPα2 KO mice. CAC was induced in these mice by intraperitoneal injection of azoxymethane followed by dextran sulfate sodium intake. Intestinal epithelial cells were isolated from colon tissues, and we performed complementary DNA microarray analysis. Cytokine expression in normal colon tissues and CAC was analyzed by quantitative polymerase chain reaction. RESULTS: Bone marrow chimeric mice showed that immune cell function between WT mice and RalGAPα2 KO mice was not significantly different in the CAC mechanism. RalGAPα2 KO mice had a significantly larger tumor number and size and a significantly higher proportion of tumors invading the submucosa than WT mice. Higher expression levels of matrix metalloproteinase-9 and matrix metalloproteinase-13 were observed in RalGAPα2 KO mice than in WT mice. The expression levels of interleukin 1ß, NLRP3, apoptosis associated speck-like protein containing a CARD, and caspase-1 were apparently increased in the tumors of RalGAPα2 KO mice compared with WT mice. NLRP3 inhibitor reduced the number of invasive tumors. CONCLUSIONS: Ral activation participates in the mechanism of CAC development via NLRP3 inflammasome activation.


Subject(s)
Colitis-Associated Neoplasms/immunology , GTPase-Activating Proteins/metabolism , Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neoplasms, Experimental/immunology , Animals , Azoxymethane/administration & dosage , Azoxymethane/toxicity , Colitis-Associated Neoplasms/chemically induced , Colitis-Associated Neoplasms/pathology , Colon/drug effects , Colon/immunology , Colon/pathology , Down-Regulation/immunology , GTPase-Activating Proteins/genetics , Humans , Inflammasomes/antagonists & inhibitors , Inflammasomes/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , ral GTP-Binding Proteins/metabolism
5.
Sci Rep ; 9(1): 5184, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30914727

ABSTRACT

The underlying therapeutic mechanism of renal tubular epithelium repair of diabetic nephropathy (DN) by bone marrow-derived mesenchymal stem cells (BM-MSCs) has not been fully elucidated. Recently, mitochondria (Mt) transfer was reported as a novel action of BM-MSCs to rescue injured cells. We investigated Mt transfer from systemically administered BM-MSCs to renal proximal tubular epithelial cells (PTECs) in streptozotocin (STZ)-induced diabetic animals. BM-MSCs also transferred their Mt to impaired PTECs when co-cultured in vitro, which suppressed apoptosis of impaired PTECs. Additionally, BM-MSC-derived isolated Mt enhanced the expression of mitochondrial superoxide dismutase 2 and Bcl-2 expression and inhibited reactive oxygen species (ROS) production in vitro. Isolated Mt also inhibited nuclear translocation of PGC-1α and restored the expression of megalin and SGLT2 under high glucose condition (HG) in PTECs. Moreover, isolated Mt directly injected under the renal capsule of STZ rats improved the cellular morphology of STZ-PTECs, and the structure of the tubular basement membrane and brush border in vivo. This study is the first to show Mt transfer from systemically administered BM-MSCs to damaged PTECs in vivo, and the first to investigate mechanisms underlying the potential therapeutic effects of Mt transfer from BM-MSCs in DN.


Subject(s)
Diabetic Nephropathies/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Kidney Tubules, Proximal/metabolism , Mesenchymal Stem Cells/metabolism , Mitochondria/metabolism , Animals , Apoptosis , Cell Line , Diabetes Mellitus, Experimental/pathology , Lectins/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sodium-Glucose Transporter 2/metabolism , Streptozocin , Superoxide Dismutase/metabolism
6.
PLoS One ; 13(9): e0204252, 2018.
Article in English | MEDLINE | ID: mdl-30240403

ABSTRACT

Increasing evidence suggests that an enriched environment (EE) ameliorates cognitive impairment by promoting repair of brain damage. However, the mechanisms by which this occurs have not been determined. To address this issue, we investigated whether an EE enhanced the capability of endogenous bone marrow-derived mesenchymal stem/stromal cells (BM-MSCs) to prevent hippocampal damage due to diabetes by focusing on miRNA carried in BM-MSC-derived exosomes. In diabetic streptozotocin (STZ) rats housed in an EE (STZ/EE), cognitive impairment was significantly reduced, and both neuronal and astroglial damage in the hippocampus was alleviated compared with STZ rats housed in conventional cages (STZ/CC). BM-MSCs isolated from STZ/CC rats had functional and morphological abnormalities that were not detected in STZ/EE BM-MSCs. The miR-146a levels in exosomes in conditioned medium of cultured BM-MSCs and serum from STZ/CC rats were decreased compared with non-diabetic rats, and the level was restored in STZ/EE rats. Thus, the data suggest that increased levels of miR-146a in sera were derived from endogenous BM-MSCs in STZ/EE rats. To examine the possibility that increased miR-146a in serum may exert anti-inflammatory effects on astrocytes in diabetic rats, astrocytes transfected with miR-146a were stimulated with advanced glycation end products (AGEs) to mimic diabetic conditions. The expression of IRAK1, NF-κB, and tumor necrosis factor-α was significantly higher in AGE-stimulated astrocytes, and these factors were decreased in miR-146a-transfected astrocytes. These results suggested that EEs stimulate up-regulation of exosomal miR-146a secretion by endogenous BM-MSCs, which exerts anti-inflammatory effects on damaged astrocytes and prevents diabetes-induced cognitive impairment.


Subject(s)
Cognitive Dysfunction/prevention & control , Diabetes Mellitus, Experimental/pathology , MicroRNAs/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Bone Marrow Cells/cytology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Cells, Cultured , Cognitive Dysfunction/etiology , Diabetes Mellitus, Experimental/complications , Exosomes/metabolism , Glycation End Products, Advanced/pharmacology , Interleukin-1 Receptor-Associated Kinases/metabolism , Male , Maze Learning , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/blood , Oxidative Stress , Rats , Transfection , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
7.
Sci Rep ; 8(1): 1161, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348535

ABSTRACT

Bone marrow-derived mesenchymal stem cells (BM-MSCs) are the most valuable source of autologous cells for transplantation and tissue regeneration to treat osteoporosis. Although BM-MSCs are the primary cells responsible for maintaining bone metabolism and homeostasis, their regenerative ability may be attenuated in postmenopausal osteoporosis patients. Therefore, we first examined potential abnormalities of BM-MSCs in an oestrogen-deficient rat model constructed by ovariectomy (OVX-MSCs). Cell proliferation, mobilisation, and regulation of osteoclasts were downregulated in OVX-MSCs. Moreover, therapeutic effects of OVX-MSCs were decreased in OVX rats. Accordingly, we developed a new activator for BM-MSCs using human umbilical cord extracts, Wharton's jelly extract supernatant (WJS), which improved cell proliferation, mobilisation and suppressive effects on activated osteoclasts in OVX-MSCs. Bone volume, RANK and TRACP expression of osteoclasts, as well as proinflammatory cytokine expression in bone tissues, were ameliorated by OVX-MSCs activated with WJS (OVX-MSCs-WJ) in OVX rats. Fusion and bone resorption activity of osteoclasts were suppressed in macrophage-induced and primary mouse bone marrow cell-induced osteoclasts via suppression of osteoclast-specific genes, such as Nfatc1, Clcn7, Atp6i and Dc-stamp, by co-culture with OVX-MSCs-WJ in vitro. In this study, we developed a new activator, WJS, which improved the functional abnormalities and therapeutic effects of BM-MSCs on postmenopausal osteoporosis.


Subject(s)
Bone Density/drug effects , Complex Mixtures/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Osteoclasts/metabolism , Osteoporosis/therapy , Umbilical Cord/chemistry , Animals , Biomarkers/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism , Coculture Techniques , Complex Mixtures/isolation & purification , Disease Models, Animal , Female , Gene Expression , Mesenchymal Stem Cells/metabolism , Mice , Osteoclasts/cytology , Osteoporosis/etiology , Osteoporosis/genetics , Osteoporosis/pathology , Ovariectomy/adverse effects , RAW 264.7 Cells , Rats , Rats, Wistar , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Tartrate-Resistant Acid Phosphatase/genetics , Tartrate-Resistant Acid Phosphatase/metabolism , Tomography, X-Ray Computed , Transcription Factors/genetics , Transcription Factors/metabolism , Wharton Jelly/chemistry
8.
Sci Rep ; 8(1): 1712, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29374250

ABSTRACT

Although the cognitive impairment in Alzheimer's disease (AD) is believed to be caused by amyloid-ß (Aß) plaques and neurofibrillary tangles (NFTs), several postmortem studies have reported cognitive normal subjects with AD brain pathology. As the mechanism underlying these discrepancies has not been clarified, we focused the neuroprotective role of astrocytes. After examining 47 donated brains, we classified brains into 3 groups, no AD pathology with no dementia (N-N), AD pathology with no dementia (AD-N), and AD pathology with dementia (AD-D), which represented 41%, 21%, and 38% of brains, respectively. No differences were found in the accumulation of Aß plaques or NFTs in the entorhinal cortex (EC) between AD-N and AD-D. Number of neurons and synaptic density were increased in AD-N compared to those in AD-D. The astrocytes in AD-N possessed longer or thicker processes, while those in AD-D possessed shorter or thinner processes in layer I/II of the EC. Astrocytes in all layers of the EC in AD-N showed enhanced GLT-1 expression in comparison to those in AD-D. Therefore these activated forms of astrocytes with increased GLT-1 expression may exert beneficial roles in preserving cognitive function, even in the presence of Aß and NFTs.


Subject(s)
Alzheimer Disease/pathology , Astrocytes/enzymology , Astrocytes/pathology , Brain/pathology , Cognition Disorders/pathology , Glutamate Plasma Membrane Transport Proteins/analysis , Aged , Aged, 80 and over , Amyloid beta-Peptides/analysis , Excitatory Amino Acid Transporter 2 , Female , Humans , Male , Neurofibrillary Tangles/pathology
9.
Sci Rep ; 7(1): 8484, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28814814

ABSTRACT

Bone marrow-derived mesenchymal stem cells (BM-MSC) has been applied as the most valuable source of autologous cell transplantation for various diseases including diabetic complications. However, hyperglycemia may cause abnormalities in intrinsic BM-MSC which might lose sufficient therapeutic effects in diabetic patients. We demonstrated the functional abnormalities in BM-MSC derived from both type 1 and type 2 diabetes models in vitro, which resulted in loss of therapeutic effects in vivo in diabetic nephropathy (DN). Then, we developed a novel method to improve abnormalities in BM-MSC using human umbilical cord extracts, namely Wharton's jelly extract supernatant (WJs). WJs is a cocktail of growth factors, extracellular matrixes and exosomes, which ameliorates proliferative capacity, motility, mitochondrial degeneration, endoplasmic reticular functions and exosome secretions in both type 1 and type 2 diabetes-derived BM-MSC (DM-MSC). Exosomes contained in WJs were a key factor for this activation, which exerted similar effects to complete WJs. DM-MSC activated by WJs ameliorated renal injury in both type 1 and type 2 DN. In this study, we developed a novel activating method using WJs to significantly increase the therapeutic effect of BM-MSC, which may allow effective autologous cell transplantation.


Subject(s)
Diabetic Nephropathies/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Bone Marrow , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Diabetic Nephropathies/pathology , Disease Models, Animal , Exosomes , Humans , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Rats, Inbred OLETF , Rats, Sprague-Dawley , Wharton Jelly/chemistry
10.
Biochem Biophys Res Commun ; 489(3): 305-311, 2017 07 29.
Article in English | MEDLINE | ID: mdl-28559141

ABSTRACT

BACKGROUND: Methods for the artificial three-dimensional (3D) culture of mouse and human small-intestinal and large-intestinal stem cells have been established with CD24+ or Paneth cell niches. In contrast, no studies have established stable 3D culture for rat colon stem cells. In this study, we established an advanced method for efficient rat colonic stem cell culture. METHODS: Using various tissue homogenates, we investigated the colonic organoid forming capacity under the TMDU protocol immediately adjacent to Ootani's 3D culture assembly in the same culture dish. Next, we examined whether the supernatant from the colon could be replaced by a colon homogenate. Finally, we identified the bioactive substances that were indispensable for efficient organoid culture using protein purification by three-step column chromatography and proteomic analysis with a quantitative nanoflow liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS: By combining Ootani's method with the TMDU protocol, we established a refined culture method for Lewis rat colon organoids, which we refer to as the modified TMDU protocol. Furthermore, we confirmed that PGE2 and galection-4 promoted rat colonic organoid formation. CONCLUSIONS: We established efficient rat colonic stem cell cultures in vitro. This success will contribute to the study of rat intestinal-disease models.


Subject(s)
Colon/cytology , Organ Culture Techniques/methods , Organoids/cytology , Organoids/growth & development , Animals , Cells, Cultured , Colon/growth & development , Rats , Stem Cells/cytology
11.
Biochem Biophys Res Commun ; 485(2): 468-475, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28192120

ABSTRACT

BACKGROUND AND AIMS: Acute graft-versus-host disease (GVHD) is a major complication after allogeneic hematopoietic stem cell transplantation, which often targets gastrointestinal (GI) tract. Osteopontin (OPN) plays an important physiological role in the efficient development of Th1 immune responses and cell survival by inhibiting apoptosis. The role of OPN in acute GI-GVHD is poorly understood. In the present study, we investigated the role of OPN in donor T cells in the pathogenicity of acute GI-GVHD. METHODS: OPN knockout (KO) mice and C57BL/6 (B6) mice were used as donors, and (C57BL/6 × DBA/2) F1 (BDF1) mice were used as allograft recipients. Mice with acute GI-GVHD were divided into three groups: the control group (BDF1→BDF1), B6 group (B6→BDF1), and OPN-KO group (OPN-KO→BDF1). Bone marrow cells and spleen cells from donors were transplanted to lethally irradiated recipients. Clinical GVHD scores were assessed daily. Recipients were euthanized on day 7 after transplantation, and colons and small intestines were collected for various analyses. RESULTS: The clinical GVHD score in the OPN-KO group was significantly increased compared with the B6 and control groups. We observed a difference in the severity of colonic GVHD between the OPN-KO group and B6 group, but not small intestinal-GVHD between these groups. Interferon-γ, Tumor necrosis factor-α, Interleukin-17A, and Interleukin-18 gene expression in the OPN-KO group was differed between the colon and small intestine. Flow cytometric analysis revealed that the fluorescence intensity of splenic and colonic CD8 T cells expressing Fas Ligand was increased in the OPN-KO group compared with the B6 group. CONCLUSION: We demonstrated that the importance of OPN in T cells in the onset of acute GI-GVHD involves regulating apoptosis of the intestinal cell via the Fas-Fas Ligand pathway.


Subject(s)
Apoptosis/immunology , Epithelial Cells/immunology , Gastrointestinal Diseases/immunology , Graft vs Host Disease/immunology , Osteopontin/immunology , Acute Disease , Allografts , Animals , Apoptosis/genetics , Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/methods , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Fas Ligand Protein/metabolism , Flow Cytometry , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/metabolism , Gene Expression/immunology , Graft vs Host Disease/etiology , Graft vs Host Disease/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lymphocyte Count , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Microscopy, Fluorescence , Osteopontin/genetics , Osteopontin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Severity of Illness Index
12.
Sci Rep ; 6: 34842, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27721418

ABSTRACT

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-ß1 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.


Subject(s)
Culture Media, Conditioned/pharmacology , Diabetic Nephropathies/pathology , Diabetic Nephropathies/therapy , Kidney Tubules/metabolism , Mesenchymal Stem Cell Transplantation/methods , Albuminuria , Animals , Bone Marrow Cells/pathology , Cells, Cultured , Cytokines/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/metabolism , Diet, High-Fat/adverse effects , Epithelial-Mesenchymal Transition/drug effects , Exosomes/metabolism , Kidney Tubules/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Rats, Inbred Lew
13.
Sci Rep ; 6: 24805, 2016 04 22.
Article in English | MEDLINE | ID: mdl-27102354

ABSTRACT

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.


Subject(s)
Astrocytes/physiology , Cognitive Dysfunction/therapy , Diabetes Complications/therapy , Exosomes/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Neurons/physiology , Administration, Intravenous , Animals , CA1 Region, Hippocampal/pathology , Disease Models, Animal , Mice , Rats , Treatment Outcome
14.
PLoS One ; 10(9): e0137801, 2015.
Article in English | MEDLINE | ID: mdl-26375822

ABSTRACT

BACKGROUND: The common disease-common variant hypothesis is insufficient to explain the complexities of Crohn's disease (CD) genetics; therefore, rare variants are expected to be important in the disease. We explored rare variants associated with susceptibility to CD in Japanese individuals by personal genomic analysis. METHODS: Two-step analyses were performed. The first step was a trio analysis with whole-exome sequence (WES) analysis and the second was a follow-up case-control association study. The WES analysis pipeline comprised Burrows-Wheeler Aligner, Picard, Genome Analysis Toolkit, and SAMTOOLS. Single nucleotide variants (SNVs)/indels were annotated and filtered by using programs implemented in ANNOVAR in combination with identity-by-descent (IBD), subsequently were subjected to the linkage based, and de novo based strategies. Finally, we conducted an association study that included 176 unrelated subjects with CD and 358 healthy control subjects. RESULTS: In family members, 234,067-297,523 SNVs/indels were detected and they were educed to 106-146 by annotation based filtering. Fifty-four CD variants common to both individuals of the affected sib pair were identified. The linkage based strategy detected five candidate variants whereas the de novo based strategy identified no variants. Consequently, five candidates were analyzed in the case-control association study. CD showed a significant association with one variant in exon 4 of IL23R, G149R [rs76418789, P = 3.9E-5, odds ratio (OR) 0.21, 95% confidence interval (CI) 0.09-0.47 for the dominant model (AA + AG versus GG), and P = 7.3E-5, OR 0.21, 95% CI 0.10-0.48 for AG versus GG, and P = 7.2E-5, OR 0.23, 95% CI 0.10-0.50 for the allele model]. CONCLUSIONS: The present study, using personal genomics analysis of a small CD pedigree, is the first to show that the low-frequency non-synonymous variant of IL23R, rs76418789, protects against CD development in Japanese subjects.


Subject(s)
Crohn Disease/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide/genetics , Precision Medicine , Receptors, Interleukin/genetics , Adult , Aged , Case-Control Studies , Crohn Disease/pathology , Female , Genomics , Humans , Male , Middle Aged , Pedigree , Young Adult
15.
J Gastroenterol ; 50(9): 962-74, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25680886

ABSTRACT

BACKGROUND: The role of mesenchymal stem/stromal cells (MSCs) in tumorigenesis remains controversial. This study aimed to determine whether heterotypic interactions between MSCs and colon cancer cells can supply contextual signals towards tumor progression. METHODS: Xenografts consisting of co-implanted human colorectal cancer cells with rat MSCs in immunodeficient mice were evaluated by tumor progression, angiogenic profiles, and MSC fate. Furthermore, we investigated how MSCs function as a cancer cell niche by co-culture experiments in vitro. RESULTS: Tumor growth progressed in two ways, either independent of or dependent on MSCs. Such cell line-specific dependency could not be explained by host immune competency. COLO 320 xenograft angiogenesis was MSC-dependent, but less dependent on vascular endothelial growth factor (VEGF), whereas HT-29 angiogenesis was not MSC-dependent, but was VEGF-dependent. MSCs and COLO 320 cells established a functional positive feedback loop that triggered formation of a cancer cell niche, leading to AKT activation. Subsequently, MSCs differentiated into pericytes that enhanced angiogenesis as a perivascular niche. In contrast, the MSC niche conferred an anti-proliferative property to HT-29 cells, through mesenchymal-epithelial transition resulting in p38 activation. CONCLUSIONS: In conclusion, MSCs demonstrate pleiotropic capabilities as a cancer cell or perivascular niche to modulate colorectal cancer cell fate in a cell line-dependent manner in a xenogeneic context.


Subject(s)
Colorectal Neoplasms/pathology , Mesenchymal Stem Cells/pathology , Neovascularization, Pathologic/pathology , Signal Transduction/physiology , Stem Cell Niche/physiology , Animals , Apoptosis , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Disease Models, Animal , Disease Progression , Heterografts , Humans , In Situ Hybridization, Fluorescence , Mice , Microarray Analysis , Neoplastic Processes , Rats , Real-Time Polymerase Chain Reaction , Transplantation, Heterologous
16.
J Gastroenterol ; 50(3): 280-6, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25618180

ABSTRACT

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.


Subject(s)
Inflammatory Bowel Diseases/therapy , Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/methods , Humans , Inflammatory Bowel Diseases/immunology , Mesenchymal Stem Cell Transplantation/methods
17.
Pain ; 155(9): 1762-1772, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24907405

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)-1ß 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-1ß 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.


Subject(s)
Amygdala/metabolism , Anxiety/etiology , Behavior, Animal/physiology , Bone Marrow/metabolism , Microglia/metabolism , Neuralgia/complications , Sciatic Nerve/injuries , Animals , Anxiety/metabolism , Chemokine CCL2/metabolism , Male , Mice , Mice, Transgenic , Neuralgia/metabolism , Neurons/metabolism
18.
Stem Cells ; 32(4): 913-25, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24715689

ABSTRACT

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 O(6)-methylguanine (O(6) MeG) adducts through O(6) MeG-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)-ß signaling because such properties were completely abrogated by absorption of TGF-ß 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-ß-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.


Subject(s)
Azoxymethane/toxicity , Carcinogens/toxicity , Colonic Neoplasms/metabolism , Mesenchymal Stem Cells/metabolism , Neoplasms, Experimental/metabolism , Animals , Cells, Cultured , Coculture Techniques , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Mesenchymal Stem Cells/pathology , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Rats , Rats, Inbred Lew , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway/drug effects
19.
Hepatology ; 59(5): 1816-29, 2014 May.
Article in English | MEDLINE | ID: mdl-24375439

ABSTRACT

UNLABELLED: 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.


Subject(s)
Bone Marrow Cells/physiology , Diabetes Mellitus, Experimental/therapy , Liver/pathology , Mesenchymal Stem Cell Transplantation , Animals , Apoptosis , Cell Communication , Cell Differentiation , Cell Lineage , Cell Movement , Cytokines/analysis , Diabetes Mellitus, Experimental/pathology , Diet, High-Fat , Insulin Resistance , Liver/metabolism , Mice , Mice, Inbred C57BL , Rats , Streptozocin
20.
J Gastroenterol ; 49(2): 270-82, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24217964

ABSTRACT

BACKGROUND: Although 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. METHODS: We 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. RESULTS: Independent 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. CONCLUSIONS: Identification 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.


Subject(s)
Colitis/drug therapy , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Apoptosis , Caco-2 Cells , Cell Cycle/drug effects , Cell Hypoxia , Cell Movement/drug effects , Cell Survival/drug effects , Chemokine CCL2/analysis , Colitis/chemically induced , Colitis/pathology , Culture Media, Conditioned/chemistry , Cytokines/analysis , Dextran Sulfate , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/physiology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Lymphocytes/drug effects , Lymphocytes/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Membrane Proteins/genetics , Mice , Oligonucleotide Array Sequence Analysis , Protein Array Analysis , Proto-Oncogene Proteins/genetics , Rats , Rats, Inbred Lew , Rats, Wistar , Trinitrobenzenesulfonic Acid , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/analysis , Wnt Proteins/genetics , Wnt Signaling Pathway/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...