Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters

Publication year range
1.
Gene Ther ; 22(12): 947-59, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26196249

ABSTRACT

Oncolytic viruses (OV) have broad potential as an adjuvant for the treatment of solid tumors. The present study addresses the feasibility of clinically applicable drugs to enhance the oncolytic potential of the OV Delta24-RGD in glioblastoma. In total, 446 drugs were screened for their viral sensitizing properties in glioblastoma stem-like cells (GSCs) in vitro. Validation was done for 10 drugs to determine synergy based on the Chou Talalay assay. Mechanistic studies were undertaken to assess viability, replication efficacy, viral infection enhancement and cell death pathway induction in a selected panel of drugs. Four viral sensitizers (fluphenazine, indirubin, lofepramine and ranolazine) were demonstrated to reproducibly synergize with Delta24-RGD in multiple assays. After validation, we underscored general applicability by testing candidate drugs in a broader context of a panel of different GSCs, various solid tumor models and multiple OVs. Overall, this study identified four viral sensitizers, which synergize with Delta24-RGD and two other strains of OVs. The viral sensitizers interact with infection, replication and cell death pathways to enhance efficacy of the OV.


Subject(s)
Glioblastoma/therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/virology , Oncolytic Virotherapy/methods , Oncolytic Viruses/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Brain Neoplasms/virology , Cell Line, Tumor , Drug Evaluation, Preclinical , Fluphenazine/pharmacology , Glioblastoma/drug therapy , Glioblastoma/virology , HCT116 Cells , Humans , Indoles/pharmacology , Oncolytic Viruses/physiology , Virus Replication/drug effects
2.
Nat Med ; 5(8): 881-7, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10426310

ABSTRACT

The occurrence of multiple tumors in an organ heralds a rapidly fatal course. Although intravascular administration may deliver oncolytic viruses/vectors to each of these tumors, its efficiency is impeded by an antiviral activity present in complement-depleted plasma of rodents and humans. Here, this activity was shown to interact with complement in a calcium-dependent fashion, and antibody neutralization studies indicated preimmune IgM has a contributing role. Short-term exposure to cyclophosphamide (CPA) partially suppressed this activity in rodents and humans. At longer time points, cyclophosphamide also abrogated neutralizing antibody responses. Cyclophosphamide treatment of rats with large single or multiple intracerebral tumors substantially increased viral survival and propagation, leading to neoplastic regression.


Subject(s)
Brain Neoplasms/immunology , Glioma/immunology , Immunosuppression Therapy , Viruses/immunology , Animals , Antibodies, Viral/blood , Antibody Formation/drug effects , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Brain Neoplasms/virology , Complement System Proteins/immunology , Cyclophosphamide/pharmacology , Female , Glioma/mortality , Glioma/therapy , Glioma/virology , Humans , Immunity, Innate/drug effects , Immunoglobulin M/blood , Male , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Rats, Nude , Survival Rate , Time Factors , Tumor Cells, Cultured , Viruses/isolation & purification
3.
J Clin Invest ; 108(2): 189-201, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11457872

ABSTRACT

A DNA nonbinding mutant of the NK2 class homeoprotein Nkx2.5 dominantly inhibits cardiogenesis in Xenopus embryos, causing a small heart to develop or blocking heart formation entirely. Recently, ten heterozygous CSX/NKX2.5 homeoprotein mutations were identified in patients with congenital atrioventricular (AV) conduction defects. All four missense mutations identified in the human homeodomain led to markedly reduced DNA binding. To examine the effect of a DNA binding-impaired mutant of mouse Csx/Nkx2.5 in the embryonic heart, we generated transgenic mice expressing one such allele, I183P, under the beta-myosin heavy chain promoter. Unexpectedly, transgenic mice were born apparently normal, but the accumulation of Csx/Nkx2.5(I183P) mutant protein in the embryo, neonate, and adult myocardium resulted in progressive and profound cardiac conduction defects and heart failure. P-R prolongation observed at 2 weeks of age rapidly progressed into complete AV block as early as 4 weeks of age. Expression of connexins 40 and 43 was dramatically decreased in the transgenic heart, which may contribute to the conduction defects in the transgenic mice. This transgenic mouse model may be useful in the study of the pathogenesis of cardiac dysfunction associated with CSX/NKX2.5 mutations in humans.


Subject(s)
Atrioventricular Node/physiopathology , Heart Defects, Congenital/physiopathology , Heart Failure/etiology , Homeodomain Proteins/metabolism , Myocardium/metabolism , Transcription Factors/metabolism , Xenopus Proteins , Age Factors , Animals , Animals, Newborn , Atrioventricular Node/abnormalities , Body Weight , Connexin 43/metabolism , Connexins/metabolism , Disease Models, Animal , Down-Regulation , Echocardiography , Female , Heart/embryology , Heart/growth & development , Heart Defects, Congenital/complications , Heart Failure/genetics , Heart Failure/metabolism , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Male , Mice , Mice, Transgenic , Mutation , Myocardium/pathology , Myocardium/ultrastructure , Organ Size , Transcription Factors/genetics , Gap Junction alpha-5 Protein
4.
Circ Res ; 88(4): 383-9, 2001 Mar 02.
Article in English | MEDLINE | ID: mdl-11230104

ABSTRACT

Although sarcomere protein gene mutations cause familial hypertrophic cardiomyopathy (FHC), individuals bearing a mutant cardiac myosin binding protein C (MyBP-C) gene usually have a better prognosis than individuals bearing beta-cardiac myosin heavy chain (MHC) gene mutations. Heterozygous mice bearing a cardiac MHC missense mutation (alphaMHC(403/+) or a cardiac MyBP-C mutation (MyBP-C(t/+)) were constructed as murine FHC models using homologous recombination in embryonic stem cells. We have compared cardiac structure and function of these mouse strains by several methods to further define mechanisms that determine the severity of FHC. Both strains demonstrated progressive left ventricular (LV) hypertrophy; however, by age 30 weeks, alphaMHC(403/+) mice demonstrated considerably more LV hypertrophy than MyBP-C(t/+) mice. In older heterozygous mice, hypertrophy continued to be more severe in the alphaMHC(403/+) mice than in the MyBP-C(t/+) mice. Consistent with this finding, hearts from 50-week-old alphaMHC(403/+) mice demonstrated increased expression of molecular markers of cardiac hypertrophy, but MyBP-C(t/+) hearts did not demonstrate expression of these molecular markers until the mice were >125 weeks old. Electrophysiological evaluation indicated that MyBP-C(t/+) mice are not as likely to have inducible ventricular tachycardia as alphaMHC(403/+) mice. In addition, cardiac function of alphaMHC(403/+) mice is significantly impaired before the development of LV hypertrophy, whereas cardiac function of MyBP-C(t/+) mice is not impaired even after the development of cardiac hypertrophy. Because these murine FHC models mimic their human counterparts, we propose that similar murine models will be useful for predicting the clinical consequences of other FHC-causing mutations. These data suggest that both electrophysiological and cardiac function studies may enable more definitive risk stratification in FHC patients.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Disease Models, Animal , Actins/genetics , Alleles , Animals , Atrial Natriuretic Factor/genetics , Blotting, Northern , Carrier Proteins/genetics , Echocardiography , Electrophysiology , Family Health , Male , Mice , Mutation , Mutation, Missense , Myocardium/chemistry , Myocardium/pathology , RNA Splicing , RNA, Messenger/metabolism , Sarcomeres/chemistry , Time Factors , Transgenes , Ventricular Dysfunction, Left
5.
Cancer Res ; 56(8): 1828-33, 1996 Apr 15.
Article in English | MEDLINE | ID: mdl-8620500

ABSTRACT

Vaccination with irradiated tumor cells genetically modified to secrete granulocyte-macrophage colony-stimulating factor (GM-CSF tumor vaccine) induces a potent systemic antitumor immunity. To develop a protocol for cancer therapy to further augment the host immune response, we examined the effects of the GM-CSF tumor vaccines simultaneously producing additional cytokines. We prepared cancer vaccines expressing double cytokines by sequential recombinant retrovirus-mediated genetic transductions. We then used a murine intracerebral tumor model in which the GM-CSF tumor vaccine was less effective in immunopotentiation and evaluated tumor vaccines producing various cytokines in conjunction with GM-CSF. The cytokine combination of GM-CSF and interleukin 4 induced more potent antitumor immunity than GM-CSF alone. An in vivo depletion test showed that CD4+, CD8+, and asialoGM1+ cells were required for the optimum function of the GM-CSF plus interleukin 4 tumor vaccine. Histological examinations revealed infiltration of inflammatory cells at the site of tumor cell challenge as well as at the site of vaccination, indicating the induction of a systemic antitumor immune response which reached the central nervous system. Our findings suggest the feasibility of applying the intensified vaccination strategy to treat human cancers including malignant brain tumors.


Subject(s)
Brain Neoplasms/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Immunotherapy , Interleukin-4/biosynthesis , Melanoma, Experimental/secondary , Vaccines, Synthetic , 3T3 Cells , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Cytokines/biosynthesis , Female , Genetic Vectors , Humans , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Retroviridae
6.
Cancer Res ; 54(22): 5757-60, 1994 Nov 15.
Article in English | MEDLINE | ID: mdl-7525049

ABSTRACT

Adoptive immunogene therapy of cancer is not widely studied, although it has been proposed as a promising strategy for cancer gene therapy. One of the major obstacles to this approach is the difficulty in introducing cytokine genes efficiently into T lymphocytes. In this report, we developed an adoptive immunotherapy model with murine tumor-specific cytotoxic T lymphocytes. By using an adenoviral vector, we achieved up to 100% gene transduction of murine T lymphocytes. Treatment of mice with the cytotoxic T lymphocytes genetically modified to produce interleukin 2 resulted in reduction of tumor metastasis and longer survival from intracerebral tumor death, providing a hopeful strategy for treatments of human cancers.


Subject(s)
Immunotherapy, Adoptive/methods , Interleukin-2/metabolism , Lymphocytes, Tumor-Infiltrating/transplantation , Melanoma, Experimental/therapy , T-Lymphocyte Subsets/transplantation , Adenoviridae/genetics , Animals , Epitopes , Female , Genetic Markers , Interleukin-2/analysis , Interleukin-2/genetics , Interleukin-7/genetics , Interleukin-7/metabolism , Lac Operon/genetics , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Transfection
7.
Cancer Res ; 57(3): 461-5, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9012474

ABSTRACT

The alpha-fetoprotein (AFP) gene is normally expressed in fetal liver and is transcriptionally silent in adult liver but overexpressed in human hepatocellular carcinoma (HCC). Here, we demonstrate that replication defective recombinant adenoviral vectors, containing the human AFP promoter/enhancer, can be used to express the Escherichia coli cytosine deaminase (CD) gene (AdAFPCD) and the beta-galactosidase gene (AdAF-PlacZ) in AFP-producing HCC cell lines. Expression of the CD gene by adenovirus from the AFP promoter/enhancer (AdAFPCD) induced cells sensitive to 5-fluorocytosine (5FC) in the AFP-producing cells but not in the AFP-nonproducing cells. Transduction by an adenoviral vector harboring an ubiquitous strong promoter and CD gene showed enzymatic activity and 5FC killing in all cell lines. When AdAFPlacZ was injected into the s.c. established hepatoma in vivo, expression of the beta-galactosidase gene was confined to AFP-producing HCC xenografts. Moreover, HCC xenografts regressed by transduction with AdAFPCD and subsequently with 5FC treatment in vivo. These findings suggest that utilization of the AFP promoter/enhancer in an adenoviral vector can confer selective expression of a heterologous suicide gene in hepatocellular carcinoma cells in vitro and in vivo.


Subject(s)
Adenoviridae/genetics , Carcinoma, Hepatocellular/therapy , Genetic Therapy , Liver Neoplasms/therapy , Nucleoside Deaminases/genetics , alpha-Fetoproteins/biosynthesis , Animals , Carcinoma, Hepatocellular/enzymology , Cytosine Deaminase , Humans , Liver Neoplasms/enzymology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Tumor Cells, Cultured
8.
Cancer Res ; 56(6): 1341-5, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8640823

ABSTRACT

We analyzed the ability of a recombinant replication-defective adenovirus vector with the carcinoembryonic antigen (CEA) promotor to transfer the thymidine kinase gene of herpes simplex virus (HSVtk) into gastric cancer cells to confer sensitivity to ganciclovir (GCV). CEA-producing gastric cancer cell lines (MKN28 and MKN45), a CEA-nonproducing gastric cancer cell line (MKN1), and a human uterine cervical cancer cell line (HeLa) were infected with a recombinant adenovirus carrying lacZ reporter gene coupled to the CEA promoter (AdCEAlacZ). The efficiency of AdCEAlacZ-mediated gene transfer was correlated with the amount of CEA produced by each cell line. Furthermore, the 50% growth inhibitory concentrations (IC50) of GCV were 21 and 5.8 microm for MKN28 and MKN45, respectively, when infected with a recombinant adenovirus carrying the HSVtk gene coupled to the CEA promoter (AdCEAtk). However, MKN1 and HeLa cells infected with AdCEAtk remained resistant to GCV (IC50 > 300 microm of GCV). In addition, a bystander killing effect was demonstrated against MKN45 cells when only 20% of AdCEAtk-infected cells were mixed with uninfected cells. These data indicate the potential for targeted gene therapy using the cell type-specific promotor of the CEA gene against gastric cancers that produce CEA.


Subject(s)
Adenoviridae/genetics , Antimetabolites, Antineoplastic/therapeutic use , Carcinoembryonic Antigen/biosynthesis , Defective Viruses/genetics , Ganciclovir/therapeutic use , Gene Transfer Techniques , Genetic Vectors/genetics , Promoter Regions, Genetic/genetics , Simplexvirus/enzymology , Stomach Neoplasms/metabolism , Stomach Neoplasms/therapy , Thymidine Kinase/genetics , Adenoviridae/enzymology , Adenoviridae/physiology , Defective Viruses/enzymology , Defective Viruses/physiology , Female , Genetic Vectors/administration & dosage , HeLa Cells/drug effects , HeLa Cells/metabolism , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Thymidine Kinase/physiology , Tumor Cells, Cultured , Uterine Neoplasms/drug therapy , Uterine Neoplasms/metabolism
9.
Cancer Res ; 47(7): 1918-23, 1987 Apr 01.
Article in English | MEDLINE | ID: mdl-3815380

ABSTRACT

The presence of estrogen-independent progesterone receptors (PgR) was demonstrated in a subline of a human endometrial cancer cell line, Ishikawa cells, although the original Ishikawa cells contained estrogen-inducible PgR. Scatchard plot analysis of cytoplasmic binding data in our subline (IK-90) revealed a high affinity binding site for R5020 (Kd, 1.0 nM) with maximum binding sites of 158 fmol/mg protein. Competition experiments showed a binding specificity similar to that of typical PgR. By low-salt sucrose gradient centrifugation, radioactive 8S and 4S peaks were found. The addition of 1 microM progesterone in culture medium resulted in a rapid nuclear translocation of cytoplasmic PgR. In contrast to the original cells, estrogen receptors could not be detected in IK-90 cells, and an addition of 17 beta-estradiol (10 nM) to culture medium failed to increase PgR. Accumulation of glycogen in cytoplasm of IK-90 cells in response to R5020 (0.1-1 microM) was observed by periodic acid-Schiff staining. The addition of R5020 to culture medium (0.1-1 microM) also caused a marked decrease in the growth of IK-90 cells, whereas the other steroids including 17 beta-estradiol, tamoxifen, testosterone, and cortisol had no significant effects. These results demonstrate for the first time the presence of a progestin-responsive human endometrial cancer cell line that contains estrogen-independent functional PgR. IK-90 cells appear to be an ideal model for studying the mechanism of the antiproliferative effect of progestin on endometrial cancer cells.


Subject(s)
Estradiol/pharmacology , Norpregnadienes/toxicity , Progesterone/toxicity , Promegestone/toxicity , Receptors, Progesterone/biosynthesis , Uterine Neoplasms/pathology , Cell Division/drug effects , Cell Line , Cell Survival/drug effects , Female , Humans , Receptors, Progesterone/drug effects , Uterine Neoplasms/metabolism
10.
Circulation ; 104(22): 2734-9, 2001 Nov 27.
Article in English | MEDLINE | ID: mdl-11723028

ABSTRACT

BACKGROUND: Homozygous mutant mice expressing a truncated form of myosin-binding protein C (MyBP-C(t/t)) develop severe dilated cardiomyopathy, whereas the heterozygous mutation (MyBP-C(t/+)) causes mild hypertrophic cardiomyopathy. Adult male MyBP-C(t/t) and MyBP-C(t/+) mice were evaluated for arrhythmia vulnerability with an in vivo electrophysiology study. METHODS AND RESULTS: Surface ECGs were obtained for heart rate, rhythm, and conduction intervals. Atrial, atrioventricular, and ventricular conduction parameters and refractoriness were assessed in 22 MyBP-C(t/t), 10 MyBP-C(t/+), and 17 wild-type MyBP-C(+/+) mice with endocardial pacing and intracardiac electrogram recording. Arrhythmia induction was attempted with standardized programmed stimulation at baseline and with isoproterenol. Heart rate variability and ambient arrhythmia activity were assessed with telemetric ECG monitors. Quantitative histological characterization was performed on serial sections of excised hearts. MyBP-C(t/t) and MyBP-C(t/+) mice have normal ECG intervals and sinus node, atrial, and ventricular conduction and refractoriness. Ventricular tachycardia was reproducibly inducible in 14 of 22 MyBP-C(t/t) mice (64%) during programmed stimulation, compared with 2 of 10 MyBP-C(t/+) mice (20%) and 0 of 17 wild-type controls (P<0.001). Ventricular ectopy was present only in MyBP-C(t/t) mice during ambulatory ECG recordings. There were no differences in heart rate variability parameters. Interstitial fibrosis correlated with genotype but did not predict arrhythmia susceptibility within the MyBP-C(t/t) group. CONCLUSIONS: MyBP-C(t/t) mice, despite prominent histopathology and ventricular dysfunction, exhibit normal conduction and refractoriness, yet are vulnerable to ventricular arrhythmias. Somatic influences between genetically identical mutant mice most likely account for variability in arrhythmia susceptibility. A sarcomeric protein gene mutation leads to a dilated cardiomyopathy and ventricular arrhythmia vulnerability phenotype.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Cardiomyopathies/physiopathology , Carrier Proteins/genetics , Heart Ventricles/physiopathology , Animals , Arrhythmias, Cardiac/complications , Arrhythmias, Cardiac/genetics , Cardiomyopathies/complications , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Disease Models, Animal , Electrocardiography , Electrophysiologic Techniques, Cardiac , Genetic Predisposition to Disease , Heart Conduction System/physiopathology , Heart Rate , Heterozygote , Homozygote , Male , Mice , Mice, Mutant Strains , Mutation , Myocardium/pathology , Phenotype , Sequence Deletion
11.
Cardiovasc Res ; 50(3): 463-73, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11376622

ABSTRACT

BACKGROUND: Atrial tachycardia and fibrillation in humans may be partly consequent to vagal stimulation. Induction of fibrillation in the small heart is considered to be impossible due to lack of a critical mass of > 100-200 mm2. Even with the recent progression of the technology of in vivo and in vitro mouse electrophysiological studies, few reports describe atrial tachycardia or fibrillation in mice. The purpose of this study was to attempt provocation of atrial tachyarrhythmia in mice using transvenous pacing following cholinergic stimulation. METHODS AND RESULTS: In vivo electrophysiology studies were performed in 14 normal mice. A six-lead ECG was recorded from surface limb leads, and an octapolar electrode catheter was inserted via jugular vein cutdown approach for simultaneous atrial and ventricular endocardial recording and pacing. Atrial tachycardia and fibrillation were inducible in one mouse at baseline electrophysiology study and eleven of fourteen mice after carbamyl choline injection. The mean duration of atrial tachycardia was 126 +/- 384 s. The longest episode lasted 35 min and only terminated after atropine injection. Reinduction of atrial tachycardia after administration of atropine was not possible. CONCLUSION: Despite the small mass of the normal mouse atria, sustained atrial tachycardia and fibrillation can be easily and reproducibly inducible with endocardial pacing after cholinergic agonist administration. This finding may contribute to our understanding of the classical theories of arrhythmogenesis and critical substrates necessary for sustaining microreentrant circuits. The techniques of transcatheter parasympathetic agonist-mediated atrial tachycardia induction may be valuable in further murine electrophysiological studies, especially mutant models with potential atrial arrhythmia phenotypes.


Subject(s)
Atrial Fibrillation/etiology , Tachycardia/etiology , Animals , Atrial Fibrillation/physiopathology , Autonomic Nervous System/physiopathology , Carbachol/pharmacology , Cardiac Pacing, Artificial , Cardiotonic Agents/pharmacology , Cholinergic Agonists/pharmacology , Disease Models, Animal , Electrocardiography , Female , Hemodynamics/drug effects , Male , Mice , Mice, Inbred C57BL , Tachycardia/physiopathology
12.
Hum Gene Ther ; 7(15): 1895-905, 1996 Oct 01.
Article in English | MEDLINE | ID: mdl-8894681

ABSTRACT

The purpose of this study was to determine the feasibility of a vaccine therapy using tumor necrosis factor (TNF) gene-transduced autologous tumor cells for the treatment of human gastrointestinal cancers, which tend to have lower immunogenicity than other cancers such as melanoma and renal cell carcinoma. We succeeded in establishing primary cultured tumor cells from 12/54 carcinomatous effusions (4 liver cancer patients, 5 gastric cancer patients, 1 pancreatic cancer patient, and 2 colon cancer patients) and in transducing the TNF gene to the tumor cells by using the retrovirus vector MFG-TNF. Even after irradiation, TNF production (0.3-3.5 U/ml per 10(6) cells per 72 hr) was confirmed for 10 of 12 transfectants, and the other two transduced cells were found to have approximately one TNF gene copy. In 7 of the 12 patients, the cytotoxic activity of killer cells to nontransduced autologous tumor cells incubated with these TNF gene transfectants was augmented. This activity was blocked with anti-HLA class I antibody or BrefeldinA (BFA), suggesting that the killer cells were cytotoxic T lymphocytes (CTL) and tumor antigens are presented with HLA class I molecules. Indeed, enhanced expression of HLA class I and/or ICAM-1 molecules on the surface of the TNF gene-transduced tumor cells were observed by fluorescence-activated cell sorting (FACS) analysis. Furthermore, natural killer (NK) and/or lymphokine-activated killer (LAK) activities determined by using K562 or Daudi cells as targets were also enhanced in some of these cases when they were incubated with TNF gene-transduced tumor cells. These findings indicate the feasibility of using TNF gene-transduced tumor cells as a vaccine in gastrointestinal cancer patients.


Subject(s)
Gastrointestinal Neoplasms/immunology , Killer Cells, Natural/immunology , Transduction, Genetic , Tumor Necrosis Factor-alpha/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gastrointestinal Neoplasms/genetics , Histocompatibility Antigens Class I/metabolism , Humans , In Vitro Techniques , Intercellular Adhesion Molecule-1/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Tumor Cells, Cultured
13.
J Invest Dermatol ; 74(4): 187-91, 1980 Apr.
Article in English | MEDLINE | ID: mdl-7373071

ABSTRACT

In order to know the distribution of testosterone 5 alpha-reductase activity in human skin, we developed a micro-method, in which we used 20-50 micrograms of various tissues microdissected from freeze-dried sections. The characteristics of this enzyme in the sebaceous gland are briefly described, as follows: the identified 5 alpha-reduced metabolites are 5 alpha-dihydrotestosterone, 5 alpha-androstane-3 beta, 17 beta-diol and 5 alpha-androstanedione; the optimal pH is about 7.5; and the apparent Km is approximately 2.4 x 10(-5) M. The measurement of 5 alpha-reductase activity of various components of the skin obtained from 7 men and 5 women revealed that the sweat gland (probably apocrine) in the axillary skin possessed the highest activity of 5 alpha-reductase: the value was nearly 400 pmoles/mg dry weight/hr in the standardized condition. The sebaceous gland also showed a high activity of 85-261 pmoles/mg/hr. The hair follicles exhibited a significantly lower activity than the sebaceous gland. The enzyme activity was negligible in the epidermis, while it was detected in the dermis though the values determined were variable probably because of contamination with other components such as sweat glands and hair follicles. Thus, the present study demonstrates that the 5 alpha-reductase activity is mainly located in the apocrine sweat gland and sebaceous gland. This suggests that 5 alpha-reduction of testosterone is an important step in mediating the action of androgens in these tissues.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Oxidoreductases/metabolism , Skin/enzymology , Adolescent , Adult , Female , Hair/enzymology , Humans , Male , Middle Aged , Sebaceous Glands/enzymology , Sweat Glands/enzymology
14.
Endocrinology ; 118(4): 1530-6, 1986 Apr.
Article in English | MEDLINE | ID: mdl-3512256

ABSTRACT

Addition of epidermal growth factor (EGF) at up to 100 ng/ml to the medium of cultured explants of mouse mammary gland increased thymidine incorporation into DNA dose dependently. Addition of retinoic acid alone at 10 micrograms/ml to the medium had no significant effect on DNA synthesis, but addition of EGF with retinoic acid enhanced the EGF-stimulated DNA synthesis. Furthermore, pretreatment of mammary explants with retinoic acid enhanced the effect of EGF plus retinoic acid on cell growth. EGF inhibited the synthesis of casein and decreased alpha-lactalbumin activity of mammary explants in culture in the presence of insulin, cortisol, and PRL. Retinoic acid alone had no significant effect on the synthesis of casein, but suppressed alpha-lactalbumin activity dose dependently. Concomitant addition of retinoic acid with EGF had no significant effect on EGF-induced inhibition of casein synthesis, but enhanced EGF-induced inhibition of alpha-lactalbumin activity dose dependently. Measurement of specific binding of [125I]EGF to mouse mammary glands in culture demonstrated that pretreatment of the explants with retinoic acid slightly, but significantly, enhanced the specific binding of EGF to its cellular receptors.


Subject(s)
Epidermal Growth Factor/pharmacology , Mammary Glands, Animal/drug effects , Tretinoin/pharmacology , Animals , Caseins/biosynthesis , Cell Survival , Cells, Cultured , DNA Replication/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Female , Hydrocortisone/pharmacology , Insulin/pharmacology , Lactalbumin/metabolism , Mammary Glands, Animal/cytology , Mice , Mice, Inbred C3H , Pregnancy , Prolactin/pharmacology , Thymidine/metabolism
15.
J Cancer Res Clin Oncol ; 121(9-10): 587-92, 1995.
Article in English | MEDLINE | ID: mdl-7559742

ABSTRACT

Irradiated tumor cells genetically modified to secrete granulocyte/macrophage-colony-stimulating factor (GM-CSF tumor vaccine) are potent stimulators of systemic antitumor immunity. For the preparation of a GM-CSF gene-modified tumor vaccine, it is important to achieve efficient genetic transduction of tumor cells, leading to an appropriate expression of the induced gene. In this report, with a view to developing a protocol for an effective cancer vaccination therapy, we examined the vaccination efficacies of tumor cells secreting GM-CSF by either adenovirus- or retrovirus-mediated genetic transduction. By using an adenoviral vector, Adex1CAmGMCSF, a highly efficient gene delivery and a high-level expression of the GM-CSF gene were achieved. Unexpectedly, animal vaccination studies showed that the GM-CSF tumor vaccine transduced with the Adex1CAmGMCSF recombinant adenovirus (adenoviral GM-CSF tumor vaccine) was less efficacious than that transduced with the MFGmGMCSF recombinant retrovirus (retroviral GM-CSF tumor vaccine). The GM-CSF serum concentration attained by the adenoviral GM-CSF tumor vaccine was much higher than that obtained by the retroviral GM-CSF tumor vaccine. Our findings indicate that an optimal level of GM-CSF production is important for the tumor vaccine to elicit an adequate response in the host antitumor immunity.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Melanoma, Experimental/prevention & control , Adenoviridae/genetics , Animals , Female , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Recombinant Proteins/administration & dosage , Retroviridae/genetics , Transduction, Genetic , Tumor Cells, Cultured , Vaccination , Vaccines/immunology
16.
Fertil Steril ; 41(2): 265-9, 1984 Feb.
Article in English | MEDLINE | ID: mdl-6698220

ABSTRACT

The specific radioimmunoassay system was developed for the titration of the antibodies to porcine zona pellucida (ZP) in human sera by using 125I-labeled purified porcine ZP as antigen, which is known to have cross-reactivity with human ZP. The antibodies in human sera were detected in 3 of 11 (27%) women with unexplained infertility, in 16 of 48 (33%) amenorrheic patients, in 4 of 12 (33%) fertile women, and in 3 of 10 (30%) men. Moreover, antibody titers in infertile women were no higher than those in fertile women and in men. These results seem to suggest that the antibodies in human sera that cross-react with porcine ZP may not be an important factor in causing infertility in women.


Subject(s)
Antibodies/analysis , Antigens , Infertility, Female/immunology , Ovum/immunology , Swine/immunology , Zona Pellucida/immunology , Animals , Cross Reactions , Female , Humans , Rabbits/immunology , Radioimmunoassay
17.
Neurosurgery ; 37(3): 478-82; discussion 483, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7501113

ABSTRACT

Female sex steroids may play a role in the proliferation of meningiomas. We investigated the progesterone receptor (PgR) immunoreactivities and the Ki-67 labeling indices in the formalin-fixed, paraffin-embedded sections of meningiomas from 39 patients. After autoclave pretreatment of the sections (which were immersed in a citrate buffer), the sections were incubated with the monoclonal antibody for the PgR and the MIB-1 monoclonal antibody for the Ki-67 antigen. In the meningiomas studied, the immunoreactivity for the PgR was moderately to strongly positive in 51%, weakly positive in 21%, and negative in 28%. The nuclear staining for the PgR was clear, and no tumors were positive for the estrogen receptor. The Ki-67 labeling indices of the PgR-positive meningiomas (mean +/- standard deviation, 2.35 +/- 2.12%) were significantly lower than those of the PgR-negative meningiomas (6.53 +/- 4.83%) (P < 0.05). Two meningiomas that had recurred more than once showed high Ki-67 labeling indices and negative immunostaining for the PgR. These findings indicate that the PgR status may be closely related to the growth potentials of the meningiomas. Our results confirm that the immunodetection of the PgR and the Ki-67 antigen on the paraffin sections of meningiomas provides a practical tool for estimating the biological behavior of the meningiomas.


Subject(s)
Biomarkers, Tumor/analysis , Meningeal Neoplasms/pathology , Meningioma/pathology , Neoplasm Proteins/analysis , Neoplasms, Hormone-Dependent/pathology , Nuclear Proteins/analysis , Receptors, Progesterone/analysis , Adult , Aged , Brain/pathology , Brain Edema/pathology , Brain Edema/surgery , Cell Division/physiology , Diagnosis, Differential , Female , Humans , Immunoenzyme Techniques , Ki-67 Antigen , Male , Meningeal Neoplasms/surgery , Meningioma/surgery , Middle Aged , Neoplasms, Hormone-Dependent/surgery , Receptors, Estrogen/analysis
18.
Int J Cardiol ; 74(2-3): 115-24, 2000 Jul 31.
Article in English | MEDLINE | ID: mdl-10962110

ABSTRACT

We examined the distributions of the activation recovery interval (ARI), which is correlated with the local action potential duration (APD), to clarify the origin of the repolarization changes in ASD. The ECGs, QRST isointegral maps and ARI isochronal maps of 21 children with ASD from 3 to 5 years old in age were studied in comparison with 21 age-matched normal children. A conventional and 87 unipolar body surface ECG were simultaneously recorded. The ARIs were determined from the first derivatives of the ECG waveforms. Abnormal ST-T patterns were observed in 11 of 21 ASD, but only in two normal children. The QRST maps of a split positive area pattern were seen in 15 of ASD but none of the normal. In the ARI maps, all the normal children exhibited a short-ARI area on the left and a long-ARI area on the right side of the chest. In 19 of ASD, the ARI distribution revealed a leftward extension of the long-ARI area on the anterior chest, a relative shortening on the right anterior chest, and a localized prolonged ARI on the left anterior chest. The results suggest that right ventricular (RV) volume overload in ASD produces a localized prolongation of the APD on the RV epicardium.


Subject(s)
Electrocardiography , Heart Septal Defects, Atrial/diagnosis , Ventricular Dysfunction, Right/diagnosis , Child, Preschool , Female , Humans , Male , Probability , Reference Values , Sensitivity and Specificity
19.
J Interv Card Electrophysiol ; 4(3): 459-67, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11046183

ABSTRACT

INTRODUCTION: Gap junctions consist of connexin (Cx) proteins that enable electrical coupling of adjacent cells and propagation of action potentials. Cx40 is solely expressed in the atrium and His-Purkinje system. The purpose of this study was to evaluate atrioventricular (AV) conduction in mice with a homozygous deletion of Connexin40 (Cx40(-/-)). METHODS: Surface ECGs, intracardiac electrophysiology (EP) studies, and ambulatory telemetry were performed in Cx40(-/-) mutant mice and wild-type (WT) controls. Atrioventricular (AV) conduction parameters and arrhythmia inducibility were evaluated using programmed stimulation. Analysis of heart rate variability was based on results of ambulatory monitoring. RESULTS: Significant findings included prolonged measures of AV refractoriness and conduction in connexin40-deficient mice, including longer PR, AH, and HV intervals, increased AV refractory periods, and increased AV Wenckebach and 2:1 block cycle lengths. Connexin40-deficient mice also had an increased incidence of inducible ventricular tachycardia, decreased basal heart rates, and increased heart rate variability. CONCLUSION: A homozygous disruption of Cx40 results in prolonged AV conduction parameters due to abnormal electrical coupling in the specialized conduction system, which may also predispose to arrhythmia vulnerability.


Subject(s)
Atrioventricular Node/physiopathology , Connexins/deficiency , Gap Junctions/metabolism , Tachycardia, Ventricular/physiopathology , Animals , Electric Stimulation , Electrocardiography , Electrodes , Electrophysiology , Heart Rate , Mice , Mice, Inbred Strains , Mice, Knockout , Mice, Mutant Strains , Models, Animal , Probability , Reference Values , Statistics, Nonparametric , Tachycardia, Ventricular/diagnosis , Tachycardia, Ventricular/etiology , Gap Junction alpha-5 Protein
20.
Clin Neurol Neurosurg ; 103(1): 13-8, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11311470

ABSTRACT

We investigated the expression of c-myc and c-sis/PDGF mRNA and protein products in 20 cases of meningiomas of various grades: 10 benign, 5 atypical and 5 anaplastic meningiomas. All cases of atypical and anaplastic meningiomas were positive for c-myc protein and mRNA by immunohistochemistry and in situ hybridisation, respectively, while all 10 benign meningiomas were negative for c-myc immunostaining, with only one benign tumour positive for c-myc mRNA. Expression of PDGF-BB protein and c-sis mRNA were seen in more than 80% of the meningioma cases and was not restricted to the histological grades of meningiomas. Semiquantitative analysis showed that the frequency of c-myc immunopositive cells positively correlated with Ki-67 proliferative indices. Our findings suggest that c-myc, but not c-sis/PDGF, has some concern to the malignancy of meningiomas.


Subject(s)
Gene Expression Regulation, Neoplastic , Meningeal Neoplasms/pathology , Meningioma/pathology , Neoplasm Proteins/analysis , Phosphoproteins/analysis , Proto-Oncogene Proteins c-myc/analysis , Proto-Oncogene Proteins c-sis/analysis , Adult , Female , Humans , Immunohistochemistry , In Situ Hybridization , Male , Meningeal Neoplasms/chemistry , Meningeal Neoplasms/classification , Meningeal Neoplasms/genetics , Meningioma/chemistry , Meningioma/classification , Meningioma/genetics , Middle Aged , Neoplasm Proteins/genetics , Phosphoproteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-sis/genetics , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL