Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
2.
Biol Pharm Bull ; 46(5): 693-699, 2023.
Article En | MEDLINE | ID: mdl-37121695

We have previously demonstrated the excellent bactericidal activity of josamycin against Staphylococcus aureus isolated from patients with atopic dermatitis (AD), with therapeutic efficacy equal to that of betamethasone. The present study was designed to evaluate the effectiveness of combination therapy with betamethasone and josamycin for AD. Betametasone (0.1%) and josamycin (0.1%) were topically administered to NC/Nga mice with severe AD-like skin lesions. Skin severity scores, histological changes in skin lesions, and serum immunoglobulin E (IgE) levels were assessed as indicators of therapeutic efficacy. Topical treatment with both drugs suppressed the skin severity score to a greater degree than betamethasone alone. This was associated with a reduction of epidermal thickening, a reduced density of dermal cellular infiltration, a decreased mast cell count in the dermis, and a reduced serum IgE level. In addition, both drugs in combination markedly reduced the expression of interferon (IFN)-γ and interleukin (IL)-4 in auricular lymph node cells, as well as the S. aureus count on the lesioned skin. These results show that simultaneous topical application of both drugs can ameliorate severe AD-like skin lesions in NC/Nga mice. It is suggested that combination therapy with betamethasone and josamycin would be beneficial for control of severe AD lesions colonized by S. aureus by inhibiting the development of both T helper (Th) type 1 (Th1) and Th2 cells and also through elimination of superficially located S. aureus.


Dermatitis, Atopic , Mice , Animals , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/pathology , Josamycin/therapeutic use , Betamethasone/therapeutic use , Staphylococcus aureus , Skin/pathology , Disease Models, Animal , Immunoglobulin E
3.
EBioMedicine ; 36: 329-346, 2018 Oct.
Article En | MEDLINE | ID: mdl-30322799

BACKGROUND: Recent large-scale clinical studies demonstrate that sodium-glucose cotransporter 2 (SGLT2) inhibitors protect the diabetic kidney. However, clinical and animal studies have not shown the changes of the total glomeruli in the whole kidney treated with SGLT2 inhibitors. METHODS: We performed computed tomography (CT) imaging on mice using synchrotron radiation to investigate the impact of luseogliflozin, a SGLT2 inhibitor, on the number and volume of glomeruli in the whole kidney. FINDINGS: We did not observe a significant difference in the total glomerular number (Nglom) among mice. Luseogliflozin redistributed the number of glomeruli in different regions, accompanied by the normalization of diabetes-augmented renal volume (Vkidney). Diabetic db/db mice had a larger glomerular volume in the mid-cortex than did control db/m mice, and luseogliflozin increased the glomerular volume in all renal cortical zones of the whole kidney in db/db mice. According to the multivariate regression analysis, hemoglobin A1c level was the most relevant determinant of Vkidney, not Nglom or mean glomerular volume (Vglom), indicating that hyperglycemia induced renal (tubular) hypertrophy, but not glomerular enlargement. Luseogliflozin increased hypoxia in the juxtamedullary region, sustained upregulated renal renin expression and plasma renin activity, and failed to decrease albuminuria by downregulating megalin in db/db mice. INTERPRETATION: Based on our findings, SGLT2 inhibitors may alter glomerular distribution and size in addition to their glucose-lowering effects, presumably by affecting oxygen metabolism and humoral factors. FUND: Funding for this research was provided by The Japan Society for the Promotion of Science, the Japan Diabetes Foundation, and Asahikawa Medical University.


Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/metabolism , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Sodium-Glucose Transporter 2/metabolism , Albuminuria , Animals , Biomarkers , Disease Models, Animal , Gene Expression , Hyperglycemia , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Mice , Organ Size , Renin/genetics , Renin/metabolism , Synchrotrons , X-Ray Microtomography
4.
Article En | MEDLINE | ID: mdl-30094053

BACKGROUND: Vascular pain is a common adverse drug reaction in colorectal cancer patients receiving peripheral venous administration of oxaliplatin. The aim of this work was to identify risk factors for vascular pain, and to examine whether currently used treatments reduce its incidence. METHODS: We conducted a multicenter retrospective study in Japanese colorectal cancer patients receiving peripheral venous administration of oxaliplatin. The effects of various treatments (administration of analgesics, addition of dexamethasone to the infusion solution for pH adjustment, dilution of the infusion solution, or use of hot gel for warming the injection site) on the incidence of vascular pain were assessed. Risk factors for vascular pain were identified by multiple logistic regression analysis. RESULTS: One hundred and ninety patients who had received an oxaliplatin-containing regimen via a peripheral venous route were analyzed. None of the preventive methods examined significantly reduced the incidence of vascular pain. BMI (BMI < 22), clinical stage (I-III) and oxaliplatin dosage (130 mg/m2 versus dose reduction) were identified as independent risk factors for development of vascular pain. The incidence of oxaliplatin-induced vascular pain was significantly higher in patients who had two or more risk factors. CONCLUSIONS: BMI, clinical stage and oxaliplatin dosage were identified as independent predictive markers for oxaliplatin-induced vascular pain. Existing treatments for vascular pain are not effective in reducing its incidence.

5.
Cancer Cell ; 33(3): 386-400.e5, 2018 03 12.
Article En | MEDLINE | ID: mdl-29478914

To identify novel targets for acute myeloid leukemia (AML) therapy, we performed genome-wide CRISPR-Cas9 screening using AML cell lines, followed by a second screen in vivo. Here, we show that the mRNA decapping enzyme scavenger (DCPS) gene is essential for AML cell survival. The DCPS enzyme interacted with components of pre-mRNA metabolic pathways, including spliceosomes, as revealed by mass spectrometry. RG3039, a DCPS inhibitor originally developed to treat spinal muscular atrophy, exhibited anti-leukemic activity via inducing pre-mRNA mis-splicing. Humans harboring germline biallelic DCPS loss-of-function mutations do not exhibit aberrant hematologic phenotypes, indicating that DCPS is dispensable for human hematopoiesis. Our findings shed light on a pre-mRNA metabolic pathway and identify DCPS as a target for AML therapy.


CRISPR-Cas Systems/drug effects , Endoribonucleases/drug effects , Leukemia/drug therapy , Muscular Atrophy, Spinal/drug therapy , Quinazolines/pharmacology , Animals , CRISPR-Cas Systems/genetics , Cell Line , Endoribonucleases/genetics , Endoribonucleases/metabolism , Humans , Leukemia/genetics , Male , Metabolic Networks and Pathways/drug effects , Mice, Inbred C57BL , Muscular Atrophy, Spinal/genetics , RNA Precursors/drug effects , RNA Precursors/genetics , RNA Splicing/drug effects , RNA Splicing/genetics , RNA, Messenger/genetics
6.
Elife ; 62017 05 29.
Article En | MEDLINE | ID: mdl-28553927

Heme is required for survival of all cells, and in most eukaryotes, is produced through a series of eight enzymatic reactions. Although heme production is critical for many cellular processes, how it is coupled to cellular differentiation is unknown. Here, using zebrafish, murine, and human models, we show that erythropoietin (EPO) signaling, together with the GATA1 transcriptional target, AKAP10, regulates heme biosynthesis during erythropoiesis at the outer mitochondrial membrane. This integrated pathway culminates with the direct phosphorylation of the crucial heme biosynthetic enzyme, ferrochelatase (FECH) by protein kinase A (PKA). Biochemical, pharmacological, and genetic inhibition of this signaling pathway result in a block in hemoglobin production and concomitant intracellular accumulation of protoporphyrin intermediates. Broadly, our results implicate aberrant PKA signaling in the pathogenesis of hematologic diseases. We propose a unifying model in which the erythroid transcriptional program works in concert with post-translational mechanisms to regulate heme metabolism during normal development.


A Kinase Anchor Proteins/metabolism , Erythropoietin/metabolism , GATA1 Transcription Factor/metabolism , Heme/biosynthesis , Signal Transduction , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Mice , Mitochondrial Membranes/metabolism , Zebrafish
7.
Science ; 351(6270): 285-9, 2016 Jan 15.
Article En | MEDLINE | ID: mdl-26816381

Genes encoding human ß-type globin undergo a developmental switch from embryonic to fetal to adult-type expression. Mutations in the adult form cause inherited hemoglobinopathies or globin disorders, including sickle cell disease and thalassemia. Some experimental results have suggested that these diseases could be treated by induction of fetal-type hemoglobin (HbF). However, the mechanisms that repress HbF in adults remain unclear. We found that the LRF/ZBTB7A transcription factor occupies fetal γ-globin genes and maintains the nucleosome density necessary for γ-globin gene silencing in adults, and that LRF confers its repressive activity through a NuRD repressor complex independent of the fetal globin repressor BCL11A. Our study may provide additional opportunities for therapeutic targeting in the treatment of hemoglobinopathies.


Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Fetal Hemoglobin/genetics , Gene Silencing , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , gamma-Globins/genetics , Anemia, Sickle Cell/genetics , Animals , Carrier Proteins/genetics , Cell Line , Chromatin/metabolism , DNA-Binding Proteins/genetics , Erythroblasts/cytology , Erythropoiesis/genetics , Humans , Mice , Mice, Knockout , Nuclear Proteins/genetics , Repressor Proteins/genetics , Sequence Deletion , Thalassemia/genetics , Transcription Factors/genetics
8.
Nat Neurosci ; 18(7): 978-87, 2015 Jul.
Article En | MEDLINE | ID: mdl-26005850

PICALM is a highly validated genetic risk factor for Alzheimer's disease (AD). We found that reduced expression of PICALM in AD and murine brain endothelium correlated with amyloid-ß (Aß) pathology and cognitive impairment. Moreover, Picalm deficiency diminished Aß clearance across the murine blood-brain barrier (BBB) and accelerated Aß pathology in a manner that was reversible by endothelial PICALM re-expression. Using human brain endothelial monolayers, we found that PICALM regulated PICALM/clathrin-dependent internalization of Aß bound to the low density lipoprotein receptor related protein-1, a key Aß clearance receptor, and guided Aß trafficking to Rab5 and Rab11, leading to Aß endothelial transcytosis and clearance. PICALM levels and Aß clearance were reduced in AD-derived endothelial monolayers, which was reversible by adenoviral-mediated PICALM transfer. Inducible pluripotent stem cell-derived human endothelial cells carrying the rs3851179 protective allele exhibited higher PICALM levels and enhanced Aß clearance. Thus, PICALM regulates Aß BBB transcytosis and clearance, which has implications for Aß brain homeostasis and clearance therapy.


Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Cerebral Cortex/metabolism , Monomeric Clathrin Assembly Proteins/metabolism , Animals , Capillaries/metabolism , Endothelium, Vascular/metabolism , Homeostasis , Humans , Metabolic Clearance Rate , Mice , Mice, Knockout , Monomeric Clathrin Assembly Proteins/deficiency , Pluripotent Stem Cells , Transcytosis
9.
Haematologica ; 100(4): 439-51, 2015 Apr.
Article En | MEDLINE | ID: mdl-25552701

Clathrin-dependent endocytosis is an essential cellular process shared by all cell types. Despite this, precisely how endocytosis is regulated in a cell-type-specific manner and how this key pathway functions physiologically or pathophysiologically remain largely unknown. PICALM, which encodes the clathrin adaptor protein PICALM, was originally identified as a component of the CALM/AF10 leukemia oncogene. Here we show, by employing a series of conditional Picalm knockout mice, that PICALM critically regulates transferrin uptake in erythroid cells by functioning as a cell-type-specific regulator of transferrin receptor endocytosis. While transferrin receptor is essential for the development of all hematopoietic lineages, Picalm was dispensable for myeloid and B-lymphoid development. Furthermore, global Picalm inactivation in adult mice did not cause gross defects in mouse fitness, except for anemia and a coat color change. Freeze-etch electron microscopy of primary erythroblasts and live-cell imaging of murine embryonic fibroblasts revealed that Picalm function is required for efficient clathrin coat maturation. We showed that the PICALM PIP2 binding domain is necessary for transferrin receptor endocytosis in erythroblasts and absolutely essential for erythroid development from mouse hematopoietic stem/progenitor cells in an erythroid culture system. We further showed that Picalm deletion entirely abrogated the disease phenotype in a Jak2(V617F) knock-in murine model of polycythemia vera. Our findings provide new insights into the regulation of cell-type-specific transferrin receptor endocytosis in vivo. They also suggest a new strategy to block cellular uptake of transferrin-bound iron, with therapeutic potential for disorders characterized by inappropriate red blood cell production, such as polycythemia vera.


Hematopoiesis/genetics , Monomeric Clathrin Assembly Proteins/genetics , Polycythemia Vera/genetics , Anemia, Hypochromic/genetics , Animals , Cell Differentiation , Clathrin-Coated Vesicles/metabolism , Clathrin-Coated Vesicles/ultrastructure , Disease Models, Animal , Endocytosis , Erythroblasts/metabolism , Erythroblasts/ultrastructure , Erythropoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunophenotyping , Lymphopoiesis/genetics , Mice , Mice, Knockout , Monomeric Clathrin Assembly Proteins/chemistry , Monomeric Clathrin Assembly Proteins/deficiency , Monomeric Clathrin Assembly Proteins/metabolism , Myelopoiesis/genetics , Phenotype , Phosphatidylinositol 4,5-Diphosphate/metabolism , Polycythemia Vera/mortality , Protein Interaction Domains and Motifs , Receptors, Transferrin/metabolism
10.
Blood ; 121(6): 918-29, 2013 Feb 07.
Article En | MEDLINE | ID: mdl-23134786

Hematopoietic stem cells (HSCs) are the most primitive cells in the hematopoietic system and are under tight regulation for self-renewal and differentiation. Notch signals are essential for the emergence of definitive hematopoiesis in mouse embryos and are critical regulators of lymphoid lineage fate determination. However, it remains unclear how Notch regulates the balance between HSC self-renewal and differentiation in the adult bone marrow (BM). Here we report a novel mechanism that prevents HSCs from undergoing premature lymphoid differentiation in BM. Using a series of in vivo mouse models and functional HSC assays, we show that leukemia/lymphoma related factor (LRF) is necessary for HSC maintenance by functioning as an erythroid-specific repressor of Delta-like 4 (Dll4) expression. Lrf deletion in erythroblasts promoted up-regulation of Dll4 in erythroblasts, sensitizing HSCs to T-cell instructive signals in the BM. Our study reveals novel cross-talk between HSCs and erythroblasts, and sheds a new light on the regulatory mechanisms regulating the balance between HSC self-renewal and differentiation.


DNA-Binding Proteins/genetics , Erythroblasts/metabolism , Hematopoietic Stem Cells/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Calcium-Binding Proteins , Cell Differentiation/genetics , Cell Proliferation , Cellular Microenvironment/genetics , DNA-Binding Proteins/metabolism , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , T-Lymphocytes/metabolism , Time Factors , Transcription Factors/metabolism , Transcriptome/genetics
11.
J Clin Invest ; 121(7): 2583-98, 2011 Jul.
Article En | MEDLINE | ID: mdl-21646720

B cells play a central role in immune system function. Deregulation of normal B cell maturation can lead to the development of autoimmune syndromes as well as B cell malignancies. Elucidation of the molecular features of normal B cell development is important for the development of new target therapies for autoimmune diseases and B cell malignancies. Employing B cell-specific conditional knockout mice, we have demonstrated here that the transcription factor leukemia/lymphoma-related factor (LRF) forms an obligate dimer in B cells and regulates mature B cell lineage fate and humoral immune responses via distinctive mechanisms. Moreover, LRF inactivation in transformed B cells attenuated their growth rate. These studies identify what we believe to be a new key factor for mature B cell development and provide a rationale for targeting LRF dimers for the treatment of autoimmune diseases and B cell malignancies.


B-Lymphocytes/immunology , DNA-Binding Proteins/immunology , Germinal Center/immunology , Transcription Factors/immunology , Animals , B-Lymphocytes/physiology , Cell Differentiation/immunology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/immunology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation , Germinal Center/cytology , Germinal Center/physiology , Mice , Mice, Knockout , Microarray Analysis , Models, Molecular , Protein Conformation , Protein Multimerization , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Spleen/cytology , Transcription Factors/chemistry , Transcription Factors/genetics
12.
Dev Cell ; 17(4): 527-40, 2009 Oct.
Article En | MEDLINE | ID: mdl-19853566

GATA-1-dependent transcription is essential for erythroid differentiation and maturation. Suppression of programmed cell death is also thought to be critical for this process; however, the link between these two features of erythropoiesis has remained elusive. Here, we show that the POZ-Krüppel family transcription factor, LRF (also known as Zbtb7a/Pokemon), is a direct target of GATA1 and plays an essential antiapoptotic role during terminal erythroid differentiation. We find that loss of Lrf leads to lethal anemia in embryos, due to increased apoptosis of late-stage erythroblasts. This programmed cell death is Arf and p53 independent and is instead mediated by upregulation of the proapoptotic factor Bim. We identify Lrf as a direct repressor of Bim transcription. In strong support of this mechanism, genetic Bim loss delays the lethality of Lrf-deficient embryos and rescues their anemia phenotype. Thus, our data define a key transcriptional cascade for effective erythropoiesis, whereby GATA-1 suppresses BIM-mediated apoptosis via LRF.


Apoptosis Regulatory Proteins/physiology , Apoptosis , DNA-Binding Proteins/physiology , Erythroblasts/cytology , GATA1 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Membrane Proteins/physiology , Proto-Oncogene Proteins/physiology , Transcription Factors/physiology , Anemia/etiology , Animals , Base Sequence , Bcl-2-Like Protein 11 , Cell Differentiation , Cell Proliferation , Cells, Cultured , Colony-Forming Units Assay , Embryonic Stem Cells/metabolism , Erythroblasts/metabolism , Erythroid Precursor Cells/metabolism , GATA1 Transcription Factor/genetics , GTP-Binding Proteins/physiology , Gene Expression Profiling , Genes, Lethal , Integrases/metabolism , Luciferases/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Mutation , Myxovirus Resistance Proteins , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Science ; 316(5826): 860-6, 2007 May 11.
Article En | MEDLINE | ID: mdl-17495164

Hematopoietic stem cells in the bone marrow give rise to lymphoid progenitors, which subsequently differentiate into B and T lymphocytes. Here we show that the proto-oncogene LRF plays an essential role in the B versus T lymphoid cell-fate decision. We demonstrate that LRF is key for instructing early lymphoid progenitors in mice to develop into B lineage cells by repressing T cell-instructive signals produced by the cell-fate signal protein, Notch. We propose a new model for lymphoid lineage commitment, in which LRF acts as a master regulator of the cell's determination of B versus T lineage.


B-Lymphocytes/cytology , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/cytology , Lymphopoiesis , Proto-Oncogenes , Receptors, Notch/metabolism , T-Lymphocytes/cytology , Transcription Factors/genetics , Animals , B-Lymphocytes/physiology , Bone Marrow Cells/cytology , Cell Lineage , Cells, Cultured , DNA-Binding Proteins/physiology , Gene Deletion , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Signal Transduction , T-Lymphocytes/physiology , Thymus Gland/cytology , Transcription Factors/physiology
...