Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Cancer Sci ; 115(2): 660-671, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38130032

ABSTRACT

REV7 is a multifunctional protein implicated in various biological processes, including DNA damage response. REV7 expression in human cancer cells affects their sensitivity to DNA-damaging agents. In the present study, we investigated the significance of REV7 in pancreatic ductal adenocarcinoma (PDAC). REV7 expression was immunohistochemically examined in 92 resected PDAC specimens and 60 endoscopic ultrasound-guided fine-needle aspiration biopsy (EUS-FNAB) specimens of unresectable PDAC treated with platinum-based chemotherapy, and its association with clinicopathologic features was analyzed. Although REV7 expression was not significantly associated with the progression of primary tumors (T-factor and Stage) in either resected or unresectable PDAC, decreased levels of REV7 expression in EUS-FNAB specimens of unresectable PDAC were significantly associated with better outcomes of platinum-based chemotherapy and a favorable prognosis. REV7-deficient PDAC cell lines showed suppressed cell growth and enhanced sensitivity to cisplatin in vitro. Tumor-bearing mice generated using REV7-deficient PDAC cell lines also showed enhanced sensitivity to cisplatin in vivo. RNA sequencing analysis using WT and REV7-deficient PDAC cell lines revealed that REV7 inactivation promoted the downregulation of genes involved in the DNA repair and the upregulation of genes involved in apoptosis. Our results indicate that decreased expression of REV7 is associated with better outcomes of platinum-based chemotherapy in PDAC by suppressing the DNA damage response. It is also suggested that REV7 is a useful biomarker for predicting the outcome of platinum-based chemotherapy and the prognosis of unresectable PDAC and is a potential target for PDAC treatment.


Subject(s)
Adenocarcinoma , Biological Phenomena , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Animals , Mice , Cisplatin/pharmacology , Cisplatin/therapeutic use , Platinum/therapeutic use , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Adenocarcinoma/drug therapy , DNA Repair/genetics
2.
Pathol Int ; 72(1): 14-24, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34637584

ABSTRACT

REV7 is a multifunctional protein implicated in DNA damage tolerance, cell cycle control, and gene expression, and is involved in the carcinogenesis of various human tumors. It has been reported that REV7 expression is associated with ultraviolet-induced mutagenesis; however, the role of REV7 expression in skin cancers, including malignant melanomas, remains unclear. In the present study, we investigated the clinical and biological significance of REV7 in malignant melanoma. Levels of REV7 expression in human skin cancers were evaluated immunohistochemically. Positive expression of REV7 was frequently observed in malignant melanomas, as well as in squamous cell carcinomas and basal cell carcinomas. Enhanced immunoreactivity to REV7 was closely linked with cell proliferation assessed by Ki-67 labeling indexes in the three skin cancers, and was related with tumor thickness in malignant melanomas. REV7 depletion in malignant melanoma cells MEWO and G361 suppressed cell proliferation, migration, and invasion abilities. REV7 depletion also affected the expression of intracellular signaling molecules AKT and ERK in MEWO cells, resulting in downregulation of ERK signal activation. In addition, REV7 depletion facilitated sensitivity to cisplatin, but not to dacarbazine, in MEWO cells. Our results suggest that REV7 expression correlates with disease progression of malignant melanoma.


Subject(s)
Mad2 Proteins/metabolism , Melanoma , Skin Neoplasms , Adolescent , Adult , Aged , Aged, 80 and over , Carcinogenesis/genetics , Cell Proliferation , Child , Child, Preschool , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , Melanoma/metabolism , Melanoma/pathology , Middle Aged , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Young Adult , Melanoma, Cutaneous Malignant
3.
Pathol Int ; 71(1): 15-23, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33112501

ABSTRACT

REV7 is involved in multiple biological processes including DNA damage tolerance, cell cycle regulation and gene expression, and is an accessory subunit of the mutation-prone DNA polymerase ζ. It has been reported that REV7 expression is associated with poor prognosis in several human cancers. The aim of this study is to investigate the significance of REV7 in lung carcinogenesis. Immunohistochemical analyses of surgically resected lung cancer specimens revealed that REV7 shows an increased expression in small cell lung carcinomas (SCLCs) when compared with other histological types of lung carcinoma. Association between REV7 expression levels and clinicopathological factors was investigated using SCLC cases with or without surgical resection. Our analyses revealed that high REV7 expression significantly correlated with tumor cell proliferation, assessed by Ki-67 labeling indices, and was negatively associated with distant metastasis and extensive-stage disease. No significant association was detected between REV7 expression and other factors, including prognosis or response to chemoradiotherapy in SCLC. Increase in REV7 expression in SCLC was confirmed using SCLC cell lines. In addition, siRNA-mediated depletion of REV7 activated the apoptotic pathway and suppressed cell growth in SCLC cells. These results suggest that REV7 plays an important role in tumor cell survival and proliferation in SCLC.


Subject(s)
Mad2 Proteins/metabolism , Small Cell Lung Carcinoma , Adult , Aged , Apoptosis , Biomarkers, Tumor , Cell Proliferation , Cell Survival , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis/pathology , Prognosis , Retrospective Studies , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology
4.
Genes Cells ; 23(7): 590-598, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29767469

ABSTRACT

Osteoporosis is a global public health problem that is increasing along with an aging population. A major determinant of osteoporosis is high bone turnover, which results from osteoclast activation. CD109 is a glycosylphosphatidylinositol-anchored glycoprotein, a deficiency that leads to a psoriasis-like skin inflammation in mice. Although the expression of CD109 has been reported in mouse pre-osteoclast cells, its function in osteoclasts in vivo remains largely unknown. To investigate the physiological role of CD109 in bone metabolism, we analyzed bones from wild-type and CD109-deficient adult mice. Micro-computed tomography analysis of the femur (thigh bone) showed that bone volume was lower in CD109-deficient mice than in wild-type mice. Bone histomorphometric analysis showed not only a reduction in bone volume but also an increase in bone turnover in CD109-deficient mice as compared with wild-type mice. Additionally, we measured serum levels of several markers of bone turnover and found a significant increase in the N-terminal telopeptide of type I collagen, a bone resorption marker, as well as alkaline phosphatase, a bone formation marker, in CD109-deficient mice. These results indicate that CD109 deficiency induces a high-turnover, osteoporosis-like phenotype, which suggests that CD109 plays a role in bone metabolism in vivo.


Subject(s)
Antigens, CD/metabolism , Bone Diseases, Metabolic/metabolism , Neoplasm Proteins/metabolism , Osteoporosis/metabolism , Animals , Biomarkers/metabolism , Bone Diseases, Metabolic/pathology , Bone Resorption , Bone and Bones/metabolism , Cell Differentiation/physiology , Collagen Type I/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/deficiency , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis , Phenotype
5.
J Cell Sci ; 126(Pt 4): 1032-45, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23264747

ABSTRACT

Chemokines have recently been reported to be involved in pathological bone destruction. However, the physiological roles of chemokines in bone metabolism in vivo have not been well documented. We analyzed the bone phenotypes in Cx3cr1-deficient mice. The mice exhibited slight but significant increases in trabecular and cortical thickness, reduced numbers of osteoclasts and increased rates of osteoid formation. Although the morphometric parameters showed marginal differences, the Cx3cr1-deficient bones showed an elevated expression of Osterix/SP7, which encodes an essential transcriptional factor for osteoblasts, whereas the gene Osteocalcin/Bglap, which encodes a late marker, was downregulated. The levels of transcripts for various osteoclastic markers, such as receptor activator of NF-κB (RANK)/TNFRSF11A, receptor activator of NF-κB ligand (RANKL)/TNFSF11, tartrate-resistant acid phosphatase 5b (TRAP5B)/ACP5B, Cathepsin K(CTSK), MMP3 and MMP13, were significantly decreased in the Cx3cr1-deficient bones. Cultured Cx3cr1-deficient osteoblastic cells showed inverse temporal patterns of osteoblastic marker expression and reduced calcium deposition. Furthermore, in vitro studies and immunofluorescence staining against CX3CR1 and CX3CL1 suggested a role for the CX3CR1-CX3CL1 axis in an early stage of osteoblast differentiation, possibly through their trans and cis interactions. Cultured Cx3cr1-deficient pre-osteoclasts showed impaired differentiation, mainly due to a deficiency of the CD115(+)CD11b(lo) osteoclastogenic population of myeloid-lineage precursors. The treatment of bone-marrow-derived osteoclastic cultures with recombinant CX3CL1 at different time points suggested that the CX3CR1-CX3CL1 axis favors the maintenance of osteoclastic precursors, but not differentiated osteoclasts. These observations uncovered novel roles of the CX3CR1-CX3CL1 axis in the differentiation of both osteoblasts and osteoclasts.


Subject(s)
Bone and Bones/cytology , Bone and Bones/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Receptors, Chemokine/metabolism , Animals , CX3C Chemokine Receptor 1 , Cells, Cultured , Flow Cytometry , Homeostasis/genetics , Homeostasis/physiology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Receptors, Chemokine/genetics
6.
Acta Cytol ; 68(2): 145-152, 2024.
Article in English | MEDLINE | ID: mdl-38555634

ABSTRACT

INTRODUCTION: Cancer genome analysis using next-generation sequencing requires adequate and high-quality DNA samples. Genomic analyses were conventionally performed using formalin-fixed paraffin-embedded sections rather than cytology samples such as cell block or smear specimens. Specimens collected from liquid-based cytology (LBC) have the potential to be sources of high-quality DNA suitable for genetic analysis even after long-term storage. METHODS: We collected breast tumor/lesion fractions from 92 residual LBC specimens using fine-needle aspiration (FNA) biopsy, including breast carcinoma (1 invasive carcinoma and 4 ductal carcinomas in situ), papillomatous lesion (5 intraductal papillomas), and fibroepithelial lesion (19 phyllodes tumors and 53 fibroadenomas) samples, and others (1 ductal adenoma, 1 hamartoma, 1 fibrocystic disease, and 7 unknown). DNA was extracted from all samples and subjected to DNA integrity number (DIN) score analysis. RESULTS: Average DIN score collected from 92 LBC specimens was significantly higher score. In addition, high-quality DNA with high DIN values (7.39 ± 0.80) was successfully extracted more than 12 months after storage of residual LBC specimens. CONCLUSION: Residual LBC specimens collected from FNA of the breast were verified to carry high-quality DNA and could serve as an alternate source for genetic analysis.


Subject(s)
Breast Neoplasms , Humans , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/diagnosis , Female , Biopsy, Fine-Needle/methods , Liquid Biopsy , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Cytodiagnosis/methods , Phyllodes Tumor/pathology , Phyllodes Tumor/genetics , Phyllodes Tumor/diagnosis , Fibroadenoma/pathology , Fibroadenoma/genetics , Fibroadenoma/diagnosis , High-Throughput Nucleotide Sequencing , Carcinoma, Intraductal, Noninfiltrating/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/diagnosis , Middle Aged , Cytology
7.
Int J Mol Sci ; 14(1): 1323-34, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23306154

ABSTRACT

Silicon quantum dots (Si-QDs) have great potential for biomedical applications, including their use as biological fluorescent markers and carriers for drug delivery systems. Biologically inert Si-QDs are less toxic than conventional cadmium-based QDs, and can modify the surface of the Si-QD with covalent bond. We synthesized water-soluble alminoprofen-conjugated Si-QDs (Ap-Si). Alminoprofen is a non-steroid anti-inflammatory drug (NSAID) used as an analgesic for rheumatism. Our results showed that the "silicon drug" is less toxic than the control Si-QD and the original drug. These phenomena indicate that the condensed surface integration of ligand/receptor-type drugs might reduce the adverse interaction between the cells and drug molecules. In addition, the medicinal effect of the Si-QDs (i.e., the inhibition of COX-2 enzyme) was maintained compared to that of the original drug. The same drug effect is related to the integration ratio of original drugs, which might control the binding interaction between COX-2 and the silicon drug. We conclude that drug conjugation with biocompatible Si-QDs is a potential method for functional pharmaceutical drug development.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Propionates/chemistry , Quantum Dots , Silicon/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arachidonic Acid/metabolism , Biocatalysis/drug effects , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprost/metabolism , Drug Evaluation, Preclinical , Enzyme-Linked Immunosorbent Assay , Hep G2 Cells , Humans , Kinetics , Microscopy, Electron, Transmission , Propionates/pharmacology , Proton Magnetic Resonance Spectroscopy , Spectroscopy, Fourier Transform Infrared
8.
J Biol Chem ; 285(37): 28826-37, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20571024

ABSTRACT

Chemokines are characterized by the homing activity of leukocytes to targeted inflammation sites. Recent research indicates that chemokines play more divergent roles in various phases of pathogenesis as well as immune reactions. The chemokine receptor, CCR1, and its ligands are thought to be involved in inflammatory bone destruction, but their physiological roles in the bone metabolism in vivo have not yet been elucidated. In the present study, we investigated the roles of CCR1 in bone metabolism using CCR1-deficient mice. Ccr1(-/-) mice have fewer and thinner trabecular bones and low mineral bone density in cancellous bones. The lack of CCR1 affects the differentiation and function of osteoblasts. Runx2, Atf4, Osteopontin, and Osteonectin were significantly up-regulated in Ccr1(-/-) mice despite sustained expression of Osterix and reduced expression of Osteocalcin, suggesting a lower potential for differentiation into mature osteoblasts. In addition, mineralized nodule formation was markedly disrupted in cultured osteoblastic cells isolated from Ccr1(-/-) mice. Osteoclastogenesis induced from cultured Ccr1(-/-) bone marrow cells yielded fewer and smaller osteoclasts due to the abrogated cell-fusion. Ccr1(-/-) osteoclasts exerted no osteolytic activity concomitant with reduced expressions of Rank and its downstream targets, implying that the defective osteoclastogenesis is involved in the bone phenotype in Ccr1(-/-) mice. The co-culture of wild-type osteoclast precursors with Ccr1(-/-) osteoblasts failed to facilitate osteoclastogenesis. This finding is most likely due to a reduction in Rankl expression. These observations suggest that the axis of CCR1 and its ligands are likely to be involved in cross-talk between osteoclasts and osteoblasts by modulating the RANK-RANKL-mediated interaction.


Subject(s)
Bone Resorption/metabolism , Cell Communication , Chemokines/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Receptors, CCR1/metabolism , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Bone Density/genetics , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Resorption/pathology , Cell Differentiation/genetics , Cells, Cultured , Chemokines/genetics , Coculture Techniques , Female , Gene Expression Regulation/genetics , Male , Mice , Mice, Knockout , Osteoblasts/pathology , Osteoclasts/pathology , Receptors, CCR1/genetics
9.
Arch Toxicol ; 85(7): 707-20, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21445587

ABSTRACT

With the development of nanotechnology, nanometer-sized products smaller than several 100 nm have been applied for all areas of science and technology. The nanometer-sized products, including carbon nanotubes, fullerene derivatives, and nanocrystals made of various materials, are widely employed as novel tools in various fields, not only in material engineering, electronics, plastics, automobile, aviation, and aerospace industries, but also even in cellular biology, molecular biology, and basic and clinical medical fields. In particular, nanocrystal quantum dots (QDs) have been widely used in biological and medical studies because of their far brighter photoemission and photostability. The physical and chemical properties of QDs have been circumstantially investigated, but little is known about the potential harmful effects of QDs on human health. In addition to the physical and chemical properties of the QDs, their toxicity and biological behavior are generally regulated by three other conditions: (1) the QD core material itself, (2) the surface modifications of the QD, and (3) the external environmental condition of the QDs. We herein report on the in vitro and in vivo toxicity and biological behavior of nanocrystals such as QDs. Accumulating evidence suggests that the QD-capping material, rather than the core metalloid complex, is responsible for the majority of their toxicity and biological activity. For example, molecules covered with a toxic agent showed cytotoxicity, whereas QDs conjugated with biomolecules retained the biological effects of the conjugate.


Subject(s)
Nanoparticles/chemistry , Nanoparticles/toxicity , Quantum Dots , Animals , Humans , Metalloids/chemistry , Metalloids/toxicity , Nanomedicine/trends , Surface Properties , Technology Transfer
10.
Intern Med ; 60(21): 3427-3433, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-33967143

ABSTRACT

We herein report a rare case of cartilage-hair hypoplasia (CHH) complicated with liver cirrhosis. A 20-year-old Japanese man with CHH was found incidentally to have liver cirrhosis and an esophageal varix. This patient had been treated for infections due to immunodeficiency since early childhood. He ultimately died of liver failure at 31 years of age. An autopsy revealed an abnormality of the interlobular bile ducts and intrahepatic cholestasis. Liver cirrhosis was thought to have been caused by chronic intrahepatic cholestasis due to biliary duct hypoplasia and changes in the intestinal microbiome. Therefore, CHH may cause biliary cirrhosis due to multiple effects.


Subject(s)
Cholestasis, Intrahepatic , Hirschsprung Disease , Primary Immunodeficiency Diseases , Adult , Cholestasis, Intrahepatic/complications , Cholestasis, Intrahepatic/diagnosis , Fatal Outcome , Hair/abnormalities , Humans , Liver Cirrhosis/complications , Male , Osteochondrodysplasias/congenital , Young Adult
11.
Nanotechnology ; 21(33): 335103, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20660952

ABSTRACT

Quantum dots (QDs) are well known for their potential application in biosensing, ex vivo live-cell imaging and in vivo animal targeting. The brain is a challenging organ for drug delivery, because the blood brain barrier (BBB) functions as a gatekeeper guarding the body from exogenous substances. Here, we evaluated the distribution of bioconjugated QDs, i.e., captopril-conjugated QDs (QDs-cap) following intraperitoneal injection into male ICR mice as a model system for determining the tissue localization of QDs, employing ICP-MS and confocal microscopy coupled with spectrometric analysis. We have demonstrated that intraperitoneally administered QDs-cap were delivered via systemic blood circulation into liver, spleen, kidney and brain at 6 h after injection. QDs-cap were located predominantly inside the blood vessels in the liver, kidney and brain, but a few were distributed in the parenchyma, especially noteworthy in the brain. Careful studies on acute as well as chronic toxicity of QDs in the brain are required prior to clinical application to humans.


Subject(s)
Brain/metabolism , Captopril/pharmacokinetics , Drug Delivery Systems/methods , Quantum Dots , Animals , Brain/blood supply , Brain/cytology , Captopril/chemistry , Immunohistochemistry , Injections, Intraperitoneal , Kidney/metabolism , Laminin , Liver/metabolism , Male , Mass Spectrometry , Mice , Mice, Inbred ICR , Microscopy, Confocal , Microscopy, Fluorescence , Tissue Distribution
12.
Int J Mol Sci ; 10(6): 2722-2732, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19582225

ABSTRACT

Mars is a CO(2)-abundant planet, whereas early Earth is thought to be also CO(2)-abundant. In addition, water was also discovered on Mars in 2008. From the facts and theory, we assumed that soda fountains were present on both planets, and this affected amino acid synthesis. Here, using a supercritical CO(2)/liquid H(2)O (10:1) system which mimicked crust soda fountains, we demonstrate production of amino acids from hydroxylamine (nitrogen source) and keto acids (oxylic acid sources). In this research, several amino acids were detected with an amino acid analyzer. Moreover, alanine polymers were detected with LC-MS. Our research lights up a new pathway in the study of life's origin.


Subject(s)
Amino Acids/chemical synthesis , Carbon Dioxide/chemistry , Water/chemistry , Amino Acids/chemistry , Hydroxylamine/chemistry , Keto Acids/chemistry , Peptides/chemistry , Temperature
13.
IEEE Trans Nanobioscience ; 6(1): 94-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17393855

ABSTRACT

Vitreous is transparent tissue located between the lens and the retina of the eye, thus, difficult to look at by even ophthalmological microscope. But vitreous is connected with some sight-threatening eye diseases, for example, retinal detachment, macular hole, epi-retinal membrane, and so forth. Quantum dots (QDs) have been applied to a wide range of biological studies by taking advantage of their fluorescence properties. We established a novel technique of aqueous colloidal QD (ACQD) as a vitreous lesion detector. When compared with some conventional dyes used for clinical situation, i.e. fluorescein, indocyanine green, and triamcinolone acetonide, ACQD exerted a higher performance to detect a Weiss Ring. Furthermore ACQD is also effective to perform vitrectomy, an eye surgery to cut and eliminate vitreous. Some functional structures in vitreous are detected clearly when ACQD was injected into an enucleated porcine eye. We demonstrated that ACQD enabled any ophthalmic surgeon to perform vitrectomy reliably, easily, and more safely. Taken together, the ACQD-oriented vitreous staining system will promote ophthalmological science, and it will raise the cure rate of eye diseases.


Subject(s)
Image Enhancement/methods , Microscopy, Fluorescence/methods , Ophthalmoscopy/methods , Quantum Dots , Vitreous Body/cytology , Animals , Humans , Staining and Labeling/methods , Swine
14.
IEEE Trans Nanobioscience ; 6(4): 341-5, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18217627

ABSTRACT

Fluorescent nanocrystal quantum dots (QDs) have been applied to a wide range of biological studies by taking advantage of their fluorescence properties. Here we show that QDs conjugated with antibody against neutrophil peroxidase, myeloperoxidase (MPO). We designed a novel method to conjugate QDs to antibody without losing any antibody function including their antigen recognizing and Fc-receptor binding activities. When we applied anti-MPO antibody (Ab) with conventional organic probes in the case of immunostaining of living cells, the antibodies lost their fluorescence because of MPO enzymic activity to produce reactive oxygen species. Our QD-conjugated anti-MPO (alpha-MPO-QDs) can detect MPO on the surface of activated neutrophils. In addition, anti-MPO-QDs did not react to the inactivated neutrophils. In conclusion, we demonstrated that antibody visualized the expression of MPO on the neutrophil surface after stimulation with proinflammatory cytokines. Taken together, these techniques have the possibility that QDs can reveal the activation of neutrophils by immunostaining and flow cytometric analysis as a powerful tool for diagnosis of the neutrophil activation in vitro.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic , Nanoparticles , Neutrophils/immunology , Peroxidase/analysis , Quantum Dots , Antibodies, Antineutrophil Cytoplasmic/administration & dosage , Antibodies, Antineutrophil Cytoplasmic/chemistry , Antigen-Antibody Complex , Antigens, Surface/analysis , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique, Direct/methods , Humans , Nanoparticles/chemistry , Neutrophil Activation , Peroxidase/immunology , Staining and Labeling
15.
Biotechnol Prog ; 23(6): 1513-6, 2007.
Article in English | MEDLINE | ID: mdl-17949107

ABSTRACT

Magnetic particles have been used widely in both biotechnological and medical fields, including for immunoassay, enzyme immobilization, drug transport, and immunological diagnosis. Especially particles with bioactive molecules such as antibodies and streptavidin are very useful tools for cell separation. Here we report affinity selection of neutrophils and macrophages from peritoneal inflammatory cells performed by thermoresponsive magnetic nanoparticles conjugated with macrophage-specific anti-F4/80 antibody. The magnetic nanoparticles, which are capped with thermoresponsive polymers, are aggregated by heating the particles over 30 degrees C and show their intrinsic magnetism. The neutrophils are concentrated approximately 90% by these magnetic nanoparticles without any activation, indicating that this novel cell separation method could fulfill a wide range of applications in analysis of the isolation of fragile cells such as neutrophils.


Subject(s)
Immunomagnetic Separation/methods , Macrophages/cytology , Nanoparticles/chemistry , Neutrophils/cytology , Animals , Flow Cytometry , Mice , Mice, Inbred C57BL , Temperature
16.
Nat Commun ; 8(1): 2226, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29263385

ABSTRACT

C-C chemokine receptor 5 (CCR5) is a co-receptor of HIV. Epidemiological findings suggest that the functional loss of CCR5 is correlated with a lower incidence of bone-destructive diseases as well as of HIV transmission. However, it is not clear whether CCR5 is involved in regulation of the function of bone cells, in addition to that of immune cells. Here we show that blockade of CCR5 using specific antibodies impairs human osteoclast function in vitro. Ccr5-deficient (Ccr5 -/- ) mice presented with dysfunctional osteoclasts and were resistant to osteoporosis induced by receptor activator of nuclear factor kappa-B ligand (RANKL), which triggers osteoporosis independently of inflammatory and immunomodulatory pathways. Furthermore, Ccr5 deficiency impairs the cellular locomotion and bone-resorption activity of osteoclasts, which is associated with the disarrangement of podosomes and adhesion complex molecules including Pyk2. Overall, the data provides evidence that CCR5 has an essential role in bone-destructive conditions through the functional regulation of osteoclasts.


Subject(s)
Bone Resorption/genetics , Cell Movement/genetics , Osteoclasts/metabolism , Osteogenesis/genetics , Osteoporosis/genetics , Receptors, CCR5/genetics , Animals , Cell Adhesion/genetics , Focal Adhesion Kinase 2/metabolism , Humans , In Vitro Techniques , Mice , Mice, Knockout , Osteoclasts/cytology , Osteoclasts/ultrastructure , Osteoporosis/chemically induced , Podosomes/ultrastructure , RANK Ligand/toxicity
17.
IEEE Trans Nanobioscience ; 5(4): 263-7, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17181025

ABSTRACT

Quantum dots (QDs) have been applied to a wide range of biological studies by taking advantage of their fluorescence properties. There is almost no method to trace small molecules including medicine. Here, we used QDs for fluorescent tracers for medicine and analyzed their kinetics and dynamics. We conjugated QDs with captopril, anti-hypertensive medicine, by an exchange reaction while retaining the medicinal properties. We investigated the medicinal effect of QD-conjugated captopril (QD-cap) in vitro and in vivo. We also evaluated the concentration and the distribution of the QD-cap in the blood and the organs with their fluorescence. We demonstrate that the QD-cap inhibits the activity of ACE in vitro. The QD-cap reduced the blood pressure of hypertensive model rats. The concentration of the QD-cap in the blood was measured by using the standard curve of the fluorescence intensity. The blood concentration of the QD-cap decrease exponentially and QD-cap has approximately the same half-life as that of captopril. In addition, the fluorescence of the QDs revealed that QD-cap accumulates in the liver, lungs, and spleen. We succeeded in analyzing the dynamics and kinetics of small molecules using fluorescence of QDs.


Subject(s)
Captopril/administration & dosage , Captopril/pharmacokinetics , Drug Evaluation, Preclinical/methods , Hypertension/drug therapy , Hypertension/metabolism , Quantum Dots , Spectrometry, Fluorescence/methods , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/pharmacokinetics , Male , Metabolic Clearance Rate , Organ Specificity , Rats , Staining and Labeling/methods , Tissue Distribution
18.
Pathog Dis ; 70(1): 28-39, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23929604

ABSTRACT

Spinal tuberculosis is a condition characterized by massive resorption of the spinal vertebrae due to the infection with Mycobacterium tuberculosis (Mtb). However, the pathogenesis of spinal tuberculosis has not been established because it was almost completely eradicated by the establishment of antibiotic treatment in the mid-20th century. In this study, we investigated the inflammatory responses of human multinucleated osteoclasts infected with virulent Mtb strain. We found that the intracellular Mtb infection of multinuclear osteoclasts resulted in the rapid growth of Mtb and an osteolytic response, rather than inflammation. In response to Mtb infection, the mononuclear osteoclast precursors produced proinflammatory cytokines including tumor necrosis factor (TNF)-α, an intrinsic characteristic they share with macrophages. In contrast, highly fused multinucleated osteoclasts incapacitated the production of these cytokines. Instead, the intracellular Mtb inside multinuclear osteoclasts escaped from the endosome/phagosome, leading to a different pattern of osteoclast activation, with the production of chemokines such as CCL5, CCL17, CCL20, CCL22, CCL24, and CCL25. Moreover, intracellular infection with an avirulent Mtb strain resulted in diminished production of these chemokines. These findings indicate that intracellular Mtb infection in multinuclear osteoclasts reprograms osteoclast development via the dysregulation of cytokines and chemokines.


Subject(s)
Chemokines/immunology , Cytokines/immunology , Mycobacterium tuberculosis/immunology , Osteoclasts/immunology , Phagosomes/immunology , Tuberculosis, Spinal/immunology , Tuberculosis, Spinal/microbiology , Endosomes/immunology , Endosomes/microbiology , Humans , Inflammation/immunology , Inflammation/microbiology , Ligands , Osteoclasts/microbiology , Phagosomes/microbiology , Tumor Necrosis Factor-alpha/immunology
19.
PLoS One ; 9(1): e83385, 2014.
Article in English | MEDLINE | ID: mdl-24400073

ABSTRACT

CD109, a glycosylphosphatidylinositol-anchored glycoprotein, is expressed at high levels in some human tumors including squamous cell carcinomas. As CD109 is reportedly cleaved by furin and its soluble form is secreted into culture medium in vitro, we hypothesized that CD109 could serve as a tumor marker in vivo. In this study, we investigated CD109 as a novel serum tumor marker using transgenic mice that overexpress mouse CD109 (mCD109-TG mice) and tumor xenografted mice inoculated with human CD109 (hCD109)-overexpressing HEK293 cells. In sera and urine of mCD109-TG mice, mCD109 was detected using western blotting. In xenografted mice, hCD109 secreted from inoculated tumors was detected in sera, using western blotting and CD109 ELISA. Concentrations of tumor-secreted CD109 increased proportionally as tumors enlarged. Concentrations of secreted CD109 decreased notably by 17 h after tumor resection, and became undetectable 48 h after resection. The half-life of tumor-secreted CD109 was about 5.86±0.17 h. These results indicate that CD109 is present in serum as a soluble form, and suggest its potential as a novel tumor marker in patients with cancers that express CD109.


Subject(s)
Antigens, CD/blood , Neoplasm Proteins/blood , Neoplasms/blood , Animals , Antigens, CD/genetics , Antigens, CD/urine , Disease Models, Animal , Female , HEK293 Cells , Heterografts , Humans , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/urine , Neoplasms/genetics , Neoplasms/pathology , Tumor Burden
20.
Bone ; 51(3): 447-58, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22766096

ABSTRACT

Confocal immunofluorescence tiling imaging revealed the spatio-temporal distributions of osterix and sclerostin in femurs from 3-day-old, 2-week-old and 4-week-old rats to be reciprocally exclusive at the tissue level. Further quantitative three-dimensional immuno fluorescence morphometry demonstrated the increasing distribution of sclerostin in the osteocytic lacuno-canalicular system specifically in diaphysis, which paralleled the cooperative participation and depletion of osterix and ß-catenin in adjacent periosteum cells. Treating MC3T3-E1 cells with BIO (a GSK3 inhibitor) induced the stabilization of ß-catenin and nuclear translocation of osterix, and negatively regulated osteocalcin/BGLAP and Dmp1. These results collectively demonstrate that the increasing distribution of sclerostin in diaphyseal cortical bone appears to be involved in the attenuation of osterix and ß-catenin in adjacent periosteum cells, thus possibly contributing to osteoblast maturation and reducing the osteoblast formation at this bone site. Our confocal microscopy-based imaging analyses provide a comprehensive and detailed view of the spatio-temporal distribution of sclerostin, ß-catenin and osterix at the tissue to subcellular level in a coherent manner, and uncovered their spatio-temporal cooperation in postnatal bone development, thus providing evidence that they link skeletogenic growth and functional bone development.


Subject(s)
Bone Development , Bone Morphogenetic Proteins/metabolism , Fluorescent Antibody Technique/methods , Imaging, Three-Dimensional/methods , Aging/metabolism , Animals , Animals, Newborn , Bone Development/drug effects , Cell Count , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Diaphyses/cytology , Diaphyses/drug effects , Diaphyses/growth & development , Diaphyses/metabolism , Femur/cytology , Femur/drug effects , Femur/growth & development , Femur/metabolism , Genetic Markers , Indoles/pharmacology , Male , Mice , Microscopy, Confocal , Osteocytes/cytology , Osteocytes/drug effects , Osteocytes/metabolism , Oximes/pharmacology , Protein Stability/drug effects , Protein Transport/drug effects , Rats , Sp7 Transcription Factor , Time Factors , Transcription Factors/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL