Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Int Heart J ; 65(2): 199-210, 2024.
Article in English | MEDLINE | ID: mdl-38556331

ABSTRACT

Infective endocarditis (IE) is a highly fatal disease in cases of delayed diagnosis and treatment, although its incidence is low. However, there have been few single-center studies in which the risk of in-hospital death from IE was stratified according to laboratory findings on admission and the organism responsible for IE. In this study, a total of 162 patients who were admitted to our hospital during the period from 2009 to 2021, who were suspected of having IE according to the modified Duke classification, and for whom IE was confirmed by transesophageal echocardiography were retrospectively analyzed. Patients were observed for a mean-period of 43.7 days with the primary endpoint being in-hospital death. The in-hospital death group had a lower level of hemoglobin (Hb), higher white blood cell (WBC) count, lower level of estimated glomerular filtration rate (eGFR), and higher frequency of Staphylococcus being the causative agent than those in the non-in-hospital death group. In overall multivariate analysis, Hb, WBC count, eGFR, and Staphylococcus as the causative agent were identified to be significant prognostic determinants. IE patients with Hb < 10.6 g/dL, WBC count > 1.4 × 104/µL, eGFR < 28.1 mL/minute/1.7 m2, and Staphylococcus as the causative agent had significantly and synergistically increased in-hospital death rates compared to those in other IE patients. Low level of Hb, high WBC count, low eGFR, and Staphylococcus as the causative agent of IE were independent predictors of in-hospital mortality, suggesting that these 4 parameters may be combined to additively stratify the risk of in-hospital mortality.


Subject(s)
Endocarditis, Bacterial , Endocarditis , Kidney Diseases , Humans , Staphylococcus , Hospital Mortality , Retrospective Studies , Endocarditis, Bacterial/diagnosis , Endocarditis/diagnosis , Leukocyte Count
2.
Am J Case Rep ; 22: e932387, 2021 Oct 09.
Article in English | MEDLINE | ID: mdl-34625525

ABSTRACT

BACKGROUND Infective endocarditis (IE) is an infectious disease that occurs in valves, centered on the endocardium and ventricular septal defects. It is a serious disease that is easily misdiagnosed and has a high mortality rate if left untreated. Edwardsiella tarda is an extremely rare cause of IE, especially in young and non-immunocompromised hosts. CASE REPORT A woman in her 20s presented to our hospital with fever of unknown cause and liver dysfunction. She was admitted to the Department of Gastroenterological Medicine owing to suspicion of gastrointestinal infection. Gastrointestinal examination, including contrast-enhanced computer tomography and endoscopic ultrasonography, was performed; however, there were no significant findings. Liver dysfunction improved spontaneously, but her fever did not improve with antibiotic treatment. Transthoracic echocardiography was performed on day 9 of hospitalization because E. tarda was detected in a blood culture test, revealing vegetation at the mitral valve. Asymptomatic cerebral infarction was shown by brain magnetic resonance imaging, and mitral valvuloplasty was performed on day 14. After surgery, transthoracic echocardiography was performed on day 22, showing no vegetation or mitral regurgitation. However, postoperative transesophageal ultrasonography performed on day 29 revealed severe mitral regurgitation. Redo mitral valvuloplasty was performed on day 38. She clinically improved and was discharged on day 67. CONCLUSIONS This is the first case in which E. tarda was diagnosed as the causative agent of IE on a native valve in a young and non-immunocompromised host. Aggressive source control resulted in a good clinical outcome.


Subject(s)
Endocarditis, Bacterial , Endocarditis , Mitral Valve Insufficiency , Edwardsiella tarda , Endocarditis, Bacterial/diagnosis , Female , Humans , Mitral Valve
3.
Oncology ; 98(3): 186-194, 2020.
Article in English | MEDLINE | ID: mdl-31846974

ABSTRACT

BACKGROUND: The clinical course of hepatocellular carcinoma (HCC) is complicated, because it often recurs and shows multiple lesions, some of which progress to a more malignant form, shortening the life of the patient. The hepatocyte growth factor receptor c-Met has been shown to play an important role in the pathogenesis of HCC, but the influence of c-Met expression on the clinical course of HCC remains to be fully elucidated. METHODS: We randomly selected and included 600 tumor specimens obtained from the primary and recurrent lesions of 319 HCC cases between 1995 and 2007. The expression of c-Met was determined by immunohistochemistry using archived formalin-fixed paraffin-embedded samples. We analyzed the correlation between c-Met expression and clinical parameters, including survival. In addition, we examined c-Met expression in the malignant transition of HCC in all cases including recurrent lesions. RESULTS: Survival analysis using the multivariate Cox proportional-regression model revealed that the prognosis was significantly better in the primary cases with high c-Met expression than in those with low c-Met expression (hazard ratio 0.159, 95% confidence interval 0.065-0.391; p < 0.001). During the course of recurrence, some cases with high c-Met expression returned to low c-Met expression. Among 40 cases with high c-Met expression, 29 survived more than 2 years after detecting the high c-Met expression. CONCLUSION: High expression of c-Met may be a prognostic factor for a good, rather than a poor, HCC prognosis. The involvement of c-Met expression in the malignant transition of recurrent HCC is obscure.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , Neoplasm Recurrence, Local , Proto-Oncogene Proteins c-met/analysis , Aged , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Disease Progression , Female , Humans , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Male , Middle Aged , Risk Factors , Time Factors , Treatment Outcome , Up-Regulation
4.
Biomed Res Int ; 2019: 5049746, 2019.
Article in English | MEDLINE | ID: mdl-30792992

ABSTRACT

The stroke-prone spontaneously hypertensive rat (SHRSP) suffers from severe hypertension and hypertensive organ damage such as cerebral stroke and kidney injury under salt-loading. By a quantitative trait locus (QTL) analysis between SHRSP and SHR (the stroke-resistant parental strain of SHRSP), two major QTLs for stroke susceptibility were identified on chromosomes 1 and 18 of SHRSP, which were confirmed in congenic strains constructed between SHRSP and SHR. As the progression of renal dysfunction was suggested to be one of the key factors inducing stroke in SHRSP, we examined effects of the stroke-related QTLs on kidney injury using two congenic strains harboring either of SHRSP-derived fragments of chromosomes 1 and 18 in the SHR genome. The congenic strains were challenged with 1% NaCl solution for 4 weeks; measurement of systolic blood pressure and urinary isoprostane level (a marker for oxidative stress) and evaluation of renal injury by quantification of genetic marker expression and histological examination were performed. We found that the congenic rats with SHRSP-derived fragment of chromosome 18 showed more severe renal damage with higher expression of Col1α-1 (a genetic marker for renal fibrosis) and higher urinary isoprostane level. In contrast, the fragment of chromosome 1 from SHRSP did not give such effects on SHR. Blood pressure was not greater in either of the congenic strains when compared with SHR. We concluded that the QTL region on chromosome 18 might deteriorate salt-induced renal injury in SHR through a blood pressure-independent mechanism.


Subject(s)
Acute Kidney Injury/genetics , Hypertension/genetics , Quantitative Trait Loci/genetics , Stroke/genetics , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Animals , Blood Pressure/genetics , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Disease Models, Animal , Humans , Hypertension/complications , Hypertension/physiopathology , Kidney/pathology , Rats , Rats, Inbred SHR/genetics , Sodium Chloride/adverse effects , Stroke/chemically induced , Stroke/complications , Stroke/pathology
5.
Hypertens Res ; 42(5): 610-617, 2019 05.
Article in English | MEDLINE | ID: mdl-30655626

ABSTRACT

Oxidative stress is involved in the pathogenesis of hypertension and hypertensive organ damage. Our previous study suggested that stroke-prone spontaneously hypertensive rats (SHRSP) exhibited greater oxidative stress than SHR and that the stroke incidence was significantly greater in SHRSP than SHR. Therefore, we hypothesized that oxidative stress was responsible for the stroke susceptibility in SHRSP. The present study constructed Prdx2 (a gene coding an antioxidative enzyme)-knockout (KO) SHR to examine whether Prdx2 knockout would make SHR more vulnerable to hypertensive organ damage, including stroke. Prdx2-KO SHR were created using CRISPR/CAS9 for genome editing. Eight-week-old male SHR and Prdx2-KO SHR were fed 1% NaCl for 2 months to induce blood pressure (BP) changes and stroke occurrence. The baseline BP was significantly greater in KO SHR, and this difference disappeared after salt loading. The life span of KO SHR was significantly reduced compared to that of SHR despite no differences in BP under salt-loading. However, no stroke was observed in KO SHR. The severity of hypertensive renal and cardiac injuries did not differ significantly between the two strains, but oxidative stress, evaluated using urinary isoprostane excretion and DHE staining, was greater in KO SHR. These results indicated that the Prdx2-depletion caused a shorter life span and modest BP increase in SHR via increased oxidative stress. The pathophysiological roles of oxidative stress in this model should be clarified in future studies.


Subject(s)
Blood Pressure , Hypertension/complications , Longevity , Peroxiredoxins/physiology , Stroke/etiology , Animals , Female , Gene Knockout Techniques , Male , Rats, Inbred SHR
6.
Cytokine ; 98: 59-70, 2017 10.
Article in English | MEDLINE | ID: mdl-28863833

ABSTRACT

T helper 2 cells produce a number of cytokines including inteleukin (IL)-5, IL-4 and IL-13. Group 2 innate lymphoid cells (ILC2s) also produce IL-5 under sterile conditions. IL-5 is interdigitating homodimeric glycoprotein and a member of the four α helical bundle motifs conserved among hematopoietic cytokines. IL-5 exerts its effects on target cells via IL-5 receptor (IL-5R), composed of an IL-5R α and ßc subunit. The membrane proximal proline-rich motif of the cytoplasmic domain of both IL-5R α and ßc subunits is essential for IL-5 signal transduction. Although IL-5 was initially identified by its ability to support the growth and terminal differentiation of mouse B cells into antibody-secreting cells, recombinant IL-5 exerts pleiotropic activities on various target cells. For example, IL-5 is now recognized as the major maturation and differentiation factor for eosinophils in mice and humans. Overexpression of IL-5 in mouse significantly increases eosinophil numbers and antibody levels in vivo, while mice lacking a functional gene for IL-5 or IL-5R display developmental and functional impairments in B cell and eosinophil lineages. In mice, the role of the IL-5/IL-5R system in the production and secretion of Immunoglobulin (Ig) M and IgA in mucosal tissues has been reported. Although eosinophils protect against invading pathogens including virus, bacteria and helminthes, they are also involved in the pathogenesis of various diseases, such as food allergy, asthma, and inflammatory bowel diseases. The recent expansion in our understanding in the context of IL-5 and IL-5-producing ILC2s in eosinophil activation and the pathogenesis of eosinophil-dependent inflammatory diseases has led to advances in therapeutic options. A new therapy currently under invetigarion in clinical trials uses humanized monoclonal antibodies against IL-5 or the IL-5R. In this review, we summarize our current understanding of the functions of IL-5 and its receptor, the innate regulation of IL-5-producing cells, and therapeutic potential of anti-IL-5 and anti-eosinophil (IL-5R) antibodies.


Subject(s)
Hypersensitivity , Interleukin-5/immunology , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Asthma/immunology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Clinical Trials as Topic , Eosinophils/immunology , Humans , Hypersensitivity/drug therapy , Hypersensitivity/immunology , Interleukin-5/antagonists & inhibitors , Interleukin-5/genetics , Lymphocytes/immunology , Mice , Receptors, Interleukin-5/immunology , Receptors, Interleukin-5/metabolism , Signal Transduction/immunology , Th2 Cells/immunology
7.
Cytotechnology ; 64(3): 249-65, 2012 May.
Article in English | MEDLINE | ID: mdl-21870215

ABSTRACT

Antibody-dependent cellular cytotoxicity (ADCC) is dependent on the fucose content of oligosaccharides bound to monoclonal antibodies (MAbs). As MAbs with a low fucose content exhibit high ADCC activity, it is important to control the defucosylation levels (deFuc%) of MAbs and to analyze the factors that affect deFuc%. In this study, we observed that the deFuc% was inversely related to culture medium osmolality for MAbs produced in the rat hybridoma cell line YB2/0, with r (2) values as high as 0.92. Moreover, deFuc% exhibited the same correlation irrespective of the type of compound used for regulating osmolality (NaCl, KCl, fucose, fructose, creatine, or mannitol) at a culture scale ranging from 1 to 400 L. We succeeded in controlling MAb deFuc% by maintaining a constant medium osmolality in both perfusion and fed-batch cultures. In agreement with these observations, reverse transcription PCR analyses revealed decreased transcription of genes involved in glycolysis, GDP-fucose supply, and fucose transfer under hypoosmotic conditions.

8.
Cytotechnology ; 63(2): 163-70, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21197574

ABSTRACT

Recently, there has been a growing demand for therapeutic monoclonal antibodies (MAbs) on the global market. Because therapeutic MAbs are more expensive than low-molecular-weight drugs, there have been strong demands to lower their production costs. Therefore, efficient methods to minimize the cost of goods are currently active areas of research. We have screened several enhancers of specific MAb production rate (SPR) using a YB2/0 cell line and found that coenzyme-Q(10) (CoQ(10)) is a promising enhancer candidate. CoQ(10) is well known as a strong antioxidant in the respiratory chain and is used for healthcare and other applications. Because CoQ(10) is negligibly water soluble, most studies are limited by low concentrations. We added CoQ(10) to a culture medium as dispersed nanoparticles at several concentrations (Q-Media) and conducted a fed-batch culture. Although the Q-Media had no effect on cumulative viable cell density, it enhanced SPR by 29%. In addition, the Q-Media had no effect on the binding or cytotoxic activity of MAbs. Q-Media also enhanced SPR with CHO and NS0 cell lines by 30%. These observations suggest that CoQ(10) serves as a powerful aid in the production of MAbs by enhancing SPR without changing the characteristics of cell growth, or adversely affecting the quality or biological activity of MAbs.

9.
J Allergy Clin Immunol ; 125(6): 1237-1244.e2, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20513521

ABSTRACT

BACKGROUND: Increased eosinophil levels have been linked to airway inflammation and asthma exacerbations. IL-5 is responsible for eosinophil differentiation, proliferation, and activation; IL-5 receptors are expressed on eosinophils and their progenitors, and targeting such receptors induces eosinophil apoptosis. OBJECTIVE: To evaluate the safety profile, pharmacokinetics, and pharmacodynamics of MEDI-563, a humanized mAb targeting the IL-5 receptor alpha chain. METHODS: Single, escalating, intravenous doses (0.0003-3 mg/kg) of MEDI-563 were administered to subjects with mild atopic asthma (n = 44) over approximately 3 to 30 minutes in this open-label study. Pulmonary function, symptom scores, adverse events, MEDI-563 pharmacokinetics, and levels of C-reactive protein (CRP), IL-6, eosinophil cationic protein (ECP), and eosinophils were evaluated. RESULTS: Mean peripheral blood (PB) eosinophil levels decreased in a dose-dependent fashion (baseline +/- SD, 0.27 +/- 0.2 x 10(3)/microL; 24 hours postdose, 0.01 +/- 0.0 x 10(3)/microL); 94.0% of subjects receiving >or=0.03 mg/kg exhibited levels between 0.00 x 10(3)/microL and 0.01 x 10(3)/microL. Eosinopenia lasted at least 8 or 12 weeks with doses of 0.03 to 0.1 and 0.3 to 3 mg/kg, respectively. ECP levels were reduced from 21.4 +/- 17.2 microg/L (baseline) to 10.3 +/- 7.0 microg/L (24 hours postdose). The most frequently reported adverse events were reduced white blood cell counts (34.1%), nasopharyngitis (27.3%), and increased blood creatine phosphokinase (25.0%). Mean C-reactive protein levels increased approximately 5.5-fold at 24 hours postdose but returned to baseline by study end; mean IL-6 levels increased approximately 3.9-fold to 4.7-fold at 6 to 12 hours postdose, respectively. Pharmacokinetic activity was dose proportional at doses of 0.03 to 3 mg/kg. CONCLUSION: Single escalating doses of MEDI-563 had an acceptable safety profile and resulted in marked reduction of PB eosinophil counts within 24 hours after dosing.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Asthma/immunology , Asthma/therapy , Eosinophils/drug effects , Recombinant Fusion Proteins/administration & dosage , Adolescent , Adult , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Apoptosis/drug effects , Apoptosis/immunology , Asthma/pathology , Asthma/physiopathology , C-Reactive Protein/metabolism , Cell Count , Eosinophil Cationic Protein/metabolism , Eosinophils/immunology , Eosinophils/metabolism , Eosinophils/pathology , Female , Follow-Up Studies , Humans , Immunotherapy , Interleukin-5 Receptor alpha Subunit/immunology , Interleukin-6/metabolism , Lymphopenia/etiology , Male , Middle Aged , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/pharmacokinetics , Respiratory Function Tests
10.
J Allergy Clin Immunol ; 125(6): 1344-1353.e2, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20513525

ABSTRACT

BACKGROUND: Peripheral blood eosinophilia and lung mucosal eosinophil infiltration are hallmarks of bronchial asthma. IL-5 is a critical cytokine for eosinophil maturation, survival, and mobilization. Attempts to target eosinophils for the treatment of asthma by means of IL-5 neutralization have only resulted in partial removal of airway eosinophils, and this warrants the development of more effective interventions to further explore the role of eosinophils in the clinical expression of asthma. OBJECTIVE: We sought to develop a novel humanized anti-IL-5 receptor alpha (IL-5Ralpha) mAb with enhanced effector function (MEDI-563) that potently depletes circulating and tissue-resident eosinophils and basophils for the treatment of asthma. METHODS: We used surface plasmon resonance to determine the binding affinity of MEDI-563 to FcgammaRIIIa. Primary human eosinophils and basophils were used to demonstrate antibody-dependent cell-mediated cytotoxicity. The binding epitope of MEDI-563 on IL-5Ralpha was determined by using site-directed mutagenesis. The consequences of MEDI-563 administration on peripheral blood and bone marrow eosinophil depletion was investigated in nonhuman primates. RESULTS: MEDI-563 binds to an epitope on IL-5Ralpha that is in close proximity to the IL-5 binding site, and it inhibits IL-5-mediated cell proliferation. MEDI-563 potently induces antibody-dependent cell-mediated cytotoxicity of both eosinophils (half-maximal effective concentration = 0.9 pmol/L) and basophils (half-maximal effective concentration = 0.5 pmol/L) in vitro. In nonhuman primates MEDI-563 depletes blood eosinophils and eosinophil precursors in the bone marrow. CONCLUSIONS: MEDI-563 might provide a novel approach for the treatment of asthma through active antibody-dependent cell-mediated depletion of eosinophils and basophils rather than through passive removal of IL-5.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Eosinophils/metabolism , Epitopes/metabolism , Interleukin-5 Receptor alpha Subunit/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Antibodies, Monoclonal/adverse effects , Antibody Affinity , Antibody-Dependent Cell Cytotoxicity , Cell Count , Eosinophils/drug effects , Eosinophils/pathology , Epitope Mapping , Female , Humans , Interleukin-5 Receptor alpha Subunit/genetics , Interleukin-5 Receptor alpha Subunit/immunology , Macaca fascicularis , Male , Mutagenesis, Site-Directed , Protein Engineering , Receptors, IgG/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Surface Plasmon Resonance
11.
Hum Antibodies ; 18(1-2): 17-27, 2009.
Article in English | MEDLINE | ID: mdl-19478395

ABSTRACT

Human interleukin-5 is the key cytokine involved in regulating the production and function of human eosinophils. IL-5 binds to its specific receptor composed of two heterogeneous alpha and beta polypeptide chains (hIL-5Ralpha and betac) that are expressed on the cell surface. The hIL-5Ralpha specifically binds IL-5 without involvement of the betac. It has been suggested that neutralizing antibodies to hIL-5Ralpha could serve as a therapeutic agent in eosinophil-associated diseases. We describe here the creation and biologic activities of a mouse monoclonal antibody against hIL-5Ralpha that blocks the following IL-5 dependent activities (a) binding of the IL-5 ligand to its receptor, (b) IL-5 dependent growth of hIL-5R expressing cells, and (c) IL-5-induced adhesion of human eosinophils. We also describe the process for humanization of the mouse Mab towards development of a therapeutic MAb. The humanized version of the monoclonal antibody also displayed potent neutralizing activity against IL-5 dependent activities.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Interleukin-5 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-5 Receptor alpha Subunit/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/pharmacology , Cell Line , Eosinophils/immunology , Humans , Immunization , Mice , Mice, Inbred BALB C , Neutralization Tests , Protein Engineering , Rats , Rats, Sprague-Dawley , Recombinant Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...