Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Adv Exp Med Biol ; 1444: 197-205, 2024.
Article in English | MEDLINE | ID: mdl-38467981

ABSTRACT

Programmed cell death-1 (PD-1) is one of the most famous coinhibitory receptors that are expressed on effector T cells to regulate their function. The PD-1 ligands, PD-L1 and PD-L2, are expressed by various cells throughout the body at steady state and their expression was further regulated within different pathological conditions such as tumor-bearing and chronic inflammatory diseases. In recent years, immune checkpoint inhibitor (ICI) therapies with anti-PD-1 or anti-PD-L1 has become a standard treatment for various malignancies and has shown remarkable antitumor effects. Since the discovery of PD-1 in 1992, a huge number of studies have been conducted to elucidate the function of PD-1. Herein, this paper provides an overview of PD-1 biological findings and sheds some light on the current technology for molecular imaging of PD-1.


Subject(s)
Neoplasms , Programmed Cell Death 1 Receptor , Humans , Programmed Cell Death 1 Receptor/metabolism , Neoplasms/metabolism , T-Lymphocytes/metabolism , B7-H1 Antigen/metabolism , Immunotherapy/methods , Molecular Imaging
2.
Nat Commun ; 14(1): 3157, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37280233

ABSTRACT

With recent advances in immune checkpoint inhibitors (ICIs), immunotherapy has become the standard treatment for various malignant tumors. Their indications and dosages have been determined empirically, taking individually conducted clinical trials into consideration, but without a standard method to evaluate them. Here we establish an advanced imaging system to visualize human PD-1 microclusters, in which a minimal T cell receptor (TCR) signaling unit co-localizes with the inhibitory co-receptor PD-1 in vitro. In these microclusters PD-1 dephosphorylates both the TCR/CD3 complex and its downstream signaling molecules via the recruitment of a phosphatase, SHP2, upon stimulation with the ligand hPD-L1. In this system, blocking antibodies for hPD-1-hPD-L1 binding inhibits hPD-1 microcluster formation, and each therapeutic antibody (pembrolizumab, nivolumab, durvalumab and atezolizumab) is characterized by a proprietary optimal concentration and combinatorial efficiency enhancement. We propose that our imaging system could digitally evaluate PD-1-mediated T cell suppression to evaluate their clinical usefulness and to develop the most suitable combinations among ICIs or between ICIs and conventional cancer treatments.


Subject(s)
Neoplasms , Programmed Cell Death 1 Receptor , Humans , Single Molecule Imaging , Nivolumab/pharmacology , Nivolumab/therapeutic use , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Receptors, Antigen, T-Cell , B7-H1 Antigen/metabolism , Immunotherapy/methods
3.
Oncol Rep ; 33(1): 292-6, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25394516

ABSTRACT

Immunotherapy has gained special attention due to its specific effects on tumor cells and systemic action to block metastasis. We recently demonstrated that ovalbumin (OVA) conjugated to the surface of nanoparticles (NPs) (OVA­NPs) can manipulate humoral immune responses. In the present study, we aimed to ascertain whether vaccination with OVA-NPs entrapping IL-7 (OVA-NPs-IL-7) are able to induce antitumor immune responses in vivo. Pretreatment with a subcutaneous inoculation of OVA-NPs delayed the growth of thymic lymphoma cells expressing a model tumor antigen OVA (E.G7-OVA), and OVA-NPs-IL-7 substantially blocked the growth of E.G7-OVA tumor cells, although NPs-IL-7 alone had a meager effect, as assessed by the mean tumor size and the percentage of tumor-free mice. However, pretreatment with OVA-NPs-IL-7 failed to reduce the growth of parental thymic tumor cells, suggesting that the antitumor effect was antigen-specific. A tetramer assay revealed that vaccination with OVA-NPs-IL-7 tended to enhance the proportion of cytotoxic T cells (CTLs) specific for OVA. When the tumor-free mice inoculated with OVA-NPs-IL-7 plus EG.7 cells were rechallenged with E.G7-OVA cells, they demonstrated reduced growth compared with that in the control mice. Thus, a single subcutaneous injection of OVA-NPs-IL-7 into mice induced tumor-specific and also memory-like immune responses, resulting in regression of tumor cells. Antigens on NPs entrapping IL-7 would be a promising carrier to develop and enhance immune responses, including humoral and cellular immunity as well as a method of drug delivery to a specific target of interest.


Subject(s)
Interleukin-7/administration & dosage , Nanocapsules/administration & dosage , Ovalbumin/administration & dosage , Thymoma/therapy , Vaccination , Animals , Cell Line, Tumor , Cell Proliferation , Cytotoxicity, Immunologic , Female , Immunologic Memory , Mice, Inbred C57BL , Ovalbumin/immunology , Ovalbumin/metabolism , Thymoma/immunology , Xenograft Model Antitumor Assays
4.
Vaccine ; 32(45): 5918-24, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25211769

ABSTRACT

There is an urgent requirement for a novel vaccine that can stimulate immune responses without unwanted toxicity, including IgE elevation. We examined whether antigen ovalbumin (OVA) conjugated to the surface of nanoparticles (NPs) (OVA-NPs) with average diameter of 110nm would serve as an immune adjuvant. When BALB/c mice were immunized with OVA-NPs, they developed sufficient levels of OVA-specific IgG1 antibody responses with low levels of IgE synthesis, representing helper T (Th)2-mediated humoral immunity. OVA-specific IgG2a and IgG2b responses (i.e., Th1-mediated immunity) were also induced by secondary immunization with OVA-NPs. As expected, immunization with OVA in alum (OVA-alum) stimulated humoral immune responses, including IgG1 and IgE antibodies, with only low levels of IgG2a/IgG2b antibodies. CD4-positive T cells from mice primed with OVA-NPs produced substantial levels of IL-21 and IL-4, comparable to those from OVA-alum group. The irradiated mice receiving OVA-NPs-primed B cells together with OVA-alum-primed T cells exhibited enhanced anti-OVA IgG2b responses relative to OVA-alum-primed B cells and T cells following stimulation with OVA-NPs. Moreover, when OVA-NPs-primed, but not OVA-alum-primed, B cells were cultured in the presence of anti-CD40 monoclonal antibody, IL-4, and IL-21, or LPS plus TGF-ß in vitro, OVA-specific IgG1 or IgG2b antibody responses were elicited, suggesting that immunization with OVA-NPs modulates B cells to generate IgG1 and IgG2b responses. Thus, OVA-NPs might exert their adjuvant action on B cells, and they represent a promising potential vaccine for generating both IgG1 and IgG2a/IgG2b antibody responses with low IgE synthesis.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alum Compounds/pharmacology , Immunoglobulin E/biosynthesis , Immunoglobulin G/immunology , Nanoparticles/administration & dosage , Ovalbumin/pharmacology , Animals , Antibody Formation , B-Lymphocytes/immunology , Cytokines/immunology , Mice, Inbred BALB C , Th1 Cells/immunology , Th2 Cells/immunology
5.
Circ J ; 77(3): 741-8, 2013.
Article in English | MEDLINE | ID: mdl-23220799

ABSTRACT

BACKGROUND: Left ventricular (LV) hypertrophy is often present in patients with diastolic heart failure. However, stiffness of hypertrophied cardiomyocytes in the transverse direction has not been fully elucidated. The aim of this study was to assess passive cardiomyocyte stiffness of hypertrophied hearts in the transverse direction and the influence of actin-myosin cross-bridge formation on the stiffness. METHODS AND RESULTS: Wistar rats received a vehicle (control) or isoproterenol (ISO) subcutaneously. After 7 days, compared with the controls, ISO administration had significantly increased heart weight and LV wall thickness and had decreased peak early annular relaxation velocity (e') assessed by echocardiography. Elastic modulus of living cardiomyocytes in the transverse direction assessed by an atomic force microscope was significantly higher in the ISO group than in controls. We added butanedione monoxime (BDM), an inhibitor of actin-myosin interaction, and blebbistatin, a specific myosin II inhibitor, to the medium. BDM and blebbistatin significantly reduced the elastic modulus of cardiomyocytes in the ISO group. X-ray diffraction analysis showed that the reflection intensity ratio (I((1,0))/I((1,1))) at diastole was not different before and after treatment with BDM, which induces complete relaxation, in control hearts, but that I((1,0))/I((1,1)) was significantly increased after BDM treatment in the ISO group, indicating residual cross-bridge formation in hypertrophied hearts. CONCLUSIONS: Passive cardiomyocyte stiffness in the transverse direction is increased in hearts with ISO-induced hypertrophy and this is caused by residual actin-myosin cross-bridge formation.


Subject(s)
Actins/metabolism , Adrenergic beta-Agonists/adverse effects , Cardiomegaly/chemically induced , Elasticity/physiology , Hypertrophy, Left Ventricular/chemically induced , Myocytes, Cardiac/pathology , Myosins/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cells, Cultured , Diacetyl/analogs & derivatives , Diacetyl/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Isoproterenol/adverse effects , Isoproterenol/pharmacology , Male , Microscopy, Atomic Force , Myocytes, Cardiac/diagnostic imaging , Myocytes, Cardiac/physiology , Organ Size/drug effects , Papillary Muscles/diagnostic imaging , Papillary Muscles/drug effects , Papillary Muscles/pathology , Radiography , Rats , Rats, Wistar , Ultrasonography
6.
Cell Mol Biol Lett ; 17(1): 36-48, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22139584

ABSTRACT

Thy28 protein is conserved among plants, bacteria, and mammalian cells. Nuclear Thy28 protein is substantially expressed in testis, liver, and immune cells such as lymphocytes. Lymphocyte apoptosis plays a crucial role in homeostasis and formation of a diverse lymphocyte repertoire. In this study, we examined whether Thy28 affects induction of apoptosis in WEHI-231 B lymphoma cells following engagement of membrane immunoglobulin (mIg). Once they were established, the Thy28-overexpressing WEHI-231 cells showed similar expression levels of IgM and class I major histocompatibility complex (MHC) molecule compared with controls. The Thy28-overexpressing cells were considerably resistant to loss of mitochondrial membrane potential (ΔΨm), caspase-3 activation, and increase in annexin-positive cells upon mIg engagement. These changes were concomitant with an increase in G1 phase associated with upregulation of p27(Kip1). The anti-IgM-induced sustained activation of c-Jun N-terminal kinase (JNK), which was associated with late-phase hydrogen peroxide (H(2)O(2)) production, was partially reduced in the Thy28-expressing cells relative to controls. Taken together, the data suggest that in WEHI-231 B lymphoma cells, Thy28 regulates mIg-mediated apoptotic events through the JNK-H(2)O(2) activation pathway, concomitant with an accumulation of cells in G1 phase associated with upregulation of p27(Kip1) in WEHI-231 B lymphoma cells.


Subject(s)
Apoptosis , Immunoglobulins/metabolism , Nuclear Proteins/metabolism , Animals , Caspase 3/metabolism , Cell Membrane/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , G1 Phase , Genes, MHC Class I/genetics , Hydrogen Peroxide/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Membrane Potential, Mitochondrial/physiology , Mice , Tumor Cells, Cultured , Up-Regulation
7.
Oncol Rep ; 22(5): 1169-72, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19787236

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) induces apoptosis in MIT6 cells derived from primary oral squamous cell carcinoma (OSCC), whereas it does not induce apoptosis in MIL6 cells derived from metastases. The present studies were performed to examine whether activation of c-Jun NH2-terminal kinase (JNK) is implicated in the differential sensitivity to TRAIL-induced apoptosis. The TRAIL-induced JNK activation in MIT6 cells was stronger than in MIL6 cells, as assessed by Western blotting using antibodies specific for phospho-JNK. To evaluate the role of JNK1 in TRAIL-induced cell death, one clone expressing the dominant-negative form of JNK1 (dnJNK1) was established. The dnJNK1-expressing cells and MIL6 cells expressed TRAIL protein at levels similar to or even greater than MIT6 cells did. When cell death was assessed by annexin V staining and mitochondrial membrane potential, kinetic studies demonstrated that the dnJNK1-expressing cells were substantially more resistant to 100 ng/ml TRAIL, comparable to MIL6 cells, at 36 and 48 h after stimulation. Collectively, the primary OSCC cell line, MIT6, is sensitive to TRAIL but its metastatic line MIL6 is resistant to TRAIL exposure. Thus, the underlying molecular mechanism of TRAIL-induced cell death involves JNK activation. These results suggest that the acquisition of TRAIL resistance provides some metastatic capacity to primary tumors.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mouth Neoplasms/metabolism , TNF-Related Apoptosis-Inducing Ligand/physiology , Blotting, Western , Drug Resistance, Neoplasm , Enzyme Activation , Flow Cytometry , Genes, Dominant , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Membrane Potential, Mitochondrial , Tumor Cells, Cultured
8.
Mol Med Rep ; 2(2): 199-203, 2009.
Article in English | MEDLINE | ID: mdl-21475813

ABSTRACT

We investigated the involvement of c-jun-N-terminal kinase (JNK) in mitochondrial depolarization and apoptosis in a human multiple myeloma cell line, U266, treated with 2-aminophenoxazine (Phx-3). It was found that, with Phx-3 administration to U266 cells, JNK was phosphorylated 2 and 7.5-fold at 6 and 24 h, respectively, compared to the Phx-3-free control. This increasing activation of JNK in U266 cells with Phx-3 correlated with cellular disorders, such as mitochondrial depolarization and cellular apoptosis. When the JNK-specific inhibitor SP6000125 was administered to the U266 cells together with Phx-3, the number of cells exhibiting mitochondrial depolarization and cellular apoptosis was significantly reduced. These results suggest that JNK activation in human multiple myeloma U266 cells may be closely associated with mitochondrial depolarization and apoptosis.

9.
J Orthop Res ; 24(6): 1153-62, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16705697

ABSTRACT

Molecular mechanisms underlying chemotherapeutic agent-induced apoptosis in sarcoma cells are not well known. Induction of apoptosis is regulated by several components including mitogen-activated protein kinases (MAPKs) comprising ERK, p38MAPKs, and c-Jun N-terminal kinase (JNK). In the present study, we examined whether activation of JNK is induced by the chemotherapeutic agents cis-diaminedichloroplatinum (cisplatin, CDDP) or doxorubicin (DXR), and whether the ectopic expression of constitutively active (MKK7-JNK1) or dominant-negative form of JNK (dnJNK) influenced apoptosis in response to the CDDP or DXR in sarcoma cell lines MG-63 and SaOS-2. The CDDP or DXR induced JNK activation in the both cell lines, as assessed by Western blotting using phosphospecific antibodies. A transient expression of the activated form of JNK sensitized the MG-63 and SaOS-2 cells to the drug-induced apoptosis, while dnJNK1 reduced the proportion of apoptotic cell death. Apoptosis was determined by flow cytometry using annexin-V Cy5. Collectively, our results indicate that JNK activation is involved in apoptotic cell death in sarcoma cell lines following stimulation with CDDP or DXR.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Doxorubicin/pharmacology , Mitogen-Activated Protein Kinase 8/biosynthesis , Sarcoma/drug therapy , Blotting, Western , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Flow Cytometry , Humans , Isoenzymes , Sarcoma/enzymology , Sarcoma/pathology
10.
Anticancer Res ; 26(2A): 1153-60, 2006.
Article in English | MEDLINE | ID: mdl-16619517

ABSTRACT

BACKGROUND: Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) in combination with a chemotherapeutic agent, cis-diammine dichloroplatinum (CDDP) or doxorubicin (DXR), has recently been demonstrated to result in enhanced apoptotic cell death in the sarcoma cell lines MG-63 and SaOS-2. DNA-damaging agents, such as CDDP induced sustained activation of c-Jun N-terminal kinase (JNK), probably leading to apoptosis. In the present study, whether JNK activation is involved in apoptotic cell death induced by combined treatment with CDDP/DXR and TRAIL was addressed. RESULTS: MG-63 or SaOS-2 cells overexpressing the dominant-negative (dn) form of JNK (dnJNK1) were established by transfection with dnJNK1 cDNA. Following stimulation with the chemotherapeutic agent CDDP or TRAIL, both MG-63 and SaOS-2 cells demonstrated enhanced cell death compared with stimulation by either agent alone, as assayed for apoptosis using annexin V staining or mitochondrial membrane potential using DiOC6 staining. Interestingly, partial inhibition of the cell death induced by the combined treatment with CDDP/DXR and TRAIL was found in MG-63 or SaOS-2 cells overexpressing dnJNK1, suggesting that JNK activation is required for the combined treatment. Moreover, induction of caspase-8 activation by TRAIL or TRAIL plus CDDP/DXR was substantially prevented by dnJNK. CONCLUSION: Efficient cell death induced by combined treatment with the chemotherapeutic agents CDDP/DXR and TRAIL is involved in JNK activation in the sarcoma cell lines MG-63 and SaOS-2. These results would be useful for treatment modalities of patients with sarcoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Sarcoma/drug therapy , Sarcoma/enzymology , Apoptosis/physiology , Apoptosis Regulatory Proteins/administration & dosage , Caspase 8 , Caspases/metabolism , Cell Line, Tumor , Cisplatin/administration & dosage , DNA Damage , Doxorubicin/administration & dosage , Enzyme Activation , Humans , Membrane Glycoproteins/administration & dosage , Membrane Potentials/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Sarcoma/pathology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/administration & dosage
11.
Oncol Rep ; 15(4): 843-8, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16525669

ABSTRACT

The aim of this study was to investigate the anticancer effects of the phenoxazine derivatives, 2-amino-4,4alpha-dihydro-4alpha,7-dimethyl-3H-phenoxazine-3-one (Phx-1), 3-amino-1,4alpha-dihydro-4alpha,8-dimethyl-2H-phenoxazine-2-one (Phx-2), and 2-aminophenoxazine-3-one (Phx-3) on human pancreatic cancer cell lines, KLM-1 and MIA-PaCa-2, in combination with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a member of the tumor necrosis factor superfamily of cytokines. Of these three phenoxazines, Phx-1 and Phx-3 inhibited proliferation of KLM-1 dose-dependently, but Phx-2 did not. Phx-3 caused both apoptosis and necrosis in KLM-1 cells, as evidenced by the phosphatidylserine externalization and propidium iodide permeable cells detected by a flow cytometric method using annexin-V and propidium iodide. Down-regulation of Bcl-2 expression appeared to be involved in the Phx-3-induced cell death. TRAIL did not affect proliferation of KLM-1, and the inhibitory effects of Phx-1 and Phx-3 on the KLM-1 cell line were not augmented by the combination with TRAIL. On the other hand, proliferation of the MIA-PaCa-2 cell line was not affected by Phx-1, Phx-2 and Phx-3, although it was significantly inhibited by TRAIL in a dose-dependent manner. Inhibitory effects of TRAIL on MIA-PaCa-2 were synergistically augmented by the addition of Phx-1 and Phx-3, but not by Phx-2. These results suggest that both Phx-1 and Phx-3 exert anticancer effects against human pancreatic cancer cells, KLM-1 and MIA-PaCa-2, through distinct action modes. Phx-1 and Phx-3 may be effective for the treatment of pancreatic cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Membrane Glycoproteins/pharmacology , Oxazines/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Humans , Molecular Structure , Oxazines/chemistry , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , TNF-Related Apoptosis-Inducing Ligand , Time Factors , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
12.
Anticancer Res ; 26(1A): 99-105, 2006.
Article in English | MEDLINE | ID: mdl-16475685

ABSTRACT

BACKGROUND: We have recently demonstrated that a truncated form of the pro-apoptotic molecule Bax-alpha (tBax) at the NH2-terminus is more potent in inducing cell death than wild-type (wt) Bax. In the present study, whether efficient cell death is induced by tBax expression from human telomerase reverse transcriptase (hTERT) promoter, which is highly active in tumor but not normal cells, was examined. MATERIALS AND METHODS: Cell death was assessed by luciferase reporter assay and the annexin staining method. RESULTS: Enforced expression of tBax resulted in cell death to a greater extent than wt Bax in two types of tumor cells: osteogenic sarcoma MG-63 and squamous cell carcinoma MIT7. The tBax sensitized these tumor cells to death induced by chemotherapeutic agents. Moreover, tBax enhanced cell death induced by the tumor necrosis factor-related apoptosis-inducing ligand to a high level, compared with wt Bax. Furthermore, tBax efficiently induced death of the MG-63 cells overexpressing Bcl-x(L), compared with wt Bax. CONCLUSION: tBax alone, or in combination with chemotherapeutic agents, would be a promising candidate for human gene therapy in the setting of carcinoma, especially for tumors containing high levels of Bcl-x(L).


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Membrane Glycoproteins/pharmacology , Neoplasms/therapy , Tumor Necrosis Factor-alpha/pharmacology , bcl-2-Associated X Protein/physiology , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Carboplatin/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cisplatin/pharmacology , DNA-Binding Proteins/genetics , Etoposide/pharmacology , Genetic Therapy/methods , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Osteosarcoma/metabolism , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Promoter Regions, Genetic , TNF-Related Apoptosis-Inducing Ligand , Telomerase/genetics , Transfection , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics , bcl-X Protein/biosynthesis
13.
Biophys J ; 86(4): 2286-94, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15041667

ABSTRACT

We studied x-ray diffraction from the left ventricular wall of an excised, perfused whole heart of a rat using x rays from the third-generation synchrotron radiation facility, SPring-8. With the beam at right angles to the long axis of the left ventricle, well-oriented, strong equatorial reflections were observed from the epicardium surface. The reflections became vertically split arcs when the beam passed through myocardium deeper in the wall, and rings were observed when the beam passed into the inner myocardium of the wall. These diffraction patterns were explained by employing a layered-spiral model of the arrangement of muscle fibers in the heart. In a quiescent heart with an expanded left ventricle, the muscle fibers at the epicardium surface were found to have a (1,0) lattice spacing smaller than in the rest of the wall. The intensity ratio of the (1,0) and (1,1) equatorial reflections decreased on contraction with a similar time course in all parts of the wall. The results show that it is possible to assign the origin of reflections in a diffraction diagram from a whole heart. This study offers a basis for interpretation of x-ray diffraction from a beating heart under physiologically and pathologically different conditions.


Subject(s)
Muscle Contraction/physiology , Muscle Fibers, Skeletal/cytology , Muscles/cytology , Myocardium/cytology , X-Ray Diffraction/methods , Animals , Heart Ventricles , Models, Theoretical , Muscle Fibers, Skeletal/chemistry , Muscles/chemistry , Myocardium/chemistry , Rats , Rats, Wistar
14.
Pflugers Arch ; 448(2): 153-60, 2004 May.
Article in English | MEDLINE | ID: mdl-14767773

ABSTRACT

We examined the sarcomere length-dependence of the spacing of the hexagonal lattice of the myofilaments and the mass transfer of myosin cross-bridges during contraction of right ventricular papillary muscle of the rat. The lattice spacing and mass transfer were measured by using X-ray diffraction, and the sarcomere length was monitored by laser diffraction at the same time. Although the lattice spacing and the sarcomere length were inversely related, their relationship was not exactly isovolumic. The cell volume decreased by about 15% when the sarcomere length was shortened from 2.3 micro m to 1.8 micro m. Twitch tension increased with sarcomere length (the Frank-Starling law). At the peak tension, the ratio of the intensity of the (1,0) equatorial reflection to that of the (1,1) reflection was smaller when the tension was greater, showing that the larger tension at a longer sarcomere length accompanies a larger amount of mass transfer of cross-bridges from the thick to the thin filament. The result suggests that the Frank-Starling law is due to an increase in the number of myosin heads attached to actin, not in the average force produced by each head.


Subject(s)
Heart/physiology , Myocardium/ultrastructure , Myosins/physiology , Sarcomeres/physiology , Sarcomeres/ultrastructure , Animals , Biophysical Phenomena , Biophysics , In Vitro Techniques , Male , Microscopy, Confocal , Myocardial Contraction/drug effects , Papillary Muscles/physiology , Papillary Muscles/ultrastructure , Rats , Rats, Wistar , X-Ray Diffraction
15.
Tissue Cell ; 35(6): 471-8, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14580360

ABSTRACT

We have recently shown that down-regulation of mouse Thy28 (mThy28) protein expression appears to be accompanied by apoptotic processes. Thymocytes from mice contain moderate amounts of mThy28 protein and undergo proliferation, differentiation, or apoptosis during murine thymic maturation. As a first step to examine the potential role of the mThy28 protein in the thymocyte development, such as positive-negative selection, the expression of mThy28 protein in the thymocyte subsets was examined. Thymocytes are separated into four subpopulations by the expression levels of CD4 and CD8: CD4-CD8- (DN), CD4+CD8+ (DP), and CD4+CD8- or CD4-CD8+ (SP). Flow cytometry analysis using three-color staining demonstrated that the mThy28 expression in immature DP cells is lower than that in DN and SP cells. The down-regulation of the mThy28 expression in the DP stage was also detected by Western blotting and reverse transcription-polymerase chain reaction (RT-PCR). The immunostaining method also showed that mThy28 protein was expressed in the medulla containing mature thymocytes, but not the cortex having immature thymocytes. The mThy28 protein in the thymocytes was mainly localized in the nucleus, as recently demonstrated in lymphoma cells, indicating that the mThy28 protein resides in the nucleus, irrespective of the cyclic or resting stage of the cell cycle. Together, the observation that mThy28 expression is down-modulated during the DP stage suggests that mThy28 protein might play some role in the positive-negative selection step in thymic maturation.


Subject(s)
Nuclear Proteins/metabolism , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Animals , Cell Lineage , Cell Nucleus/chemistry , Down-Regulation , Male , Mice , Mice, Inbred DBA , Nuclear Proteins/analysis , Nuclear Proteins/immunology , T-Lymphocyte Subsets/cytology , Thymus Gland/growth & development , Thymus Gland/immunology
16.
Melanoma Res ; 13(5): 457-64, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14512787

ABSTRACT

Malignant melanoma cells have been reported to be highly resistant to chemotherapeutic agents. To gain insight into the molecular mechanisms underlying chemotherapeutic drug resistance, we examined the role of the Bcl-2 family members Bcl-2 and Bax in cell death in the melanoma cell line G361 following stimulation with cisplatin (CDDP) or dacarbazine (DTIC). Trypan blue dye exclusion showed that both CDDP and DTIC induced death of G361 cells. Apoptotic and necrotic cell death could be distinguished by flow cytometry using combined staining with annexin V and 7-amino-actinomycin D (7-AAD). CDDP-induced cell death at a low concentration (0.6 micro g/ml) was mainly due to apoptosis (annexin V+/7-AAD-), while a mixture of apoptosis and secondary necrosis (annexin V+/7-AAD+) was found at a high concentration (6 micro g/ml). DTIC at the concentrations used induced only apoptosis. CDDP-induced apoptosis and secondary necrosis were accompanied by activation of caspase-3 and modulation of Bcl-2 family members Bcl-2 and Bax. On Western blotting Bax was seen to be upregulated with concomitant downregulation of Bcl-2. Flow cytometry, which enables measurement of protein at the single-cell level, revealed that Bcl-2+/Bax- cells were decreased, with a slight concomitant rise in Bcl-2-/Bax+ cells on stimulation with CDDP. These findings suggest that the chemotherapeutic agents CDDP and DTIC induce apoptosis and/or secondary necrosis depending on dose, probably involving the modulation of Bcl-2 family proteins.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Dactinomycin/analogs & derivatives , Melanoma/pathology , Necrosis , Proto-Oncogene Proteins c-bcl-2/metabolism , Annexin A5/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Blotting, Western , Caspase 3 , Caspases/metabolism , Cell Death , Cell Line, Tumor , Cell Survival , Cisplatin/pharmacology , Coloring Agents/pharmacology , Dacarbazine/pharmacology , Dactinomycin/pharmacology , Dose-Response Relationship, Drug , Down-Regulation , Enzyme Activation , Flow Cytometry , Fluorescent Dyes/pharmacology , Humans , Time Factors , Trypan Blue/pharmacology , Up-Regulation
17.
Cancer Lett ; 189(2): 221-30, 2003 Jan 28.
Article in English | MEDLINE | ID: mdl-12490315

ABSTRACT

Wild type (wt) p21 Bax was cleaved to generate p18 Bax during apoptotic processes by calpain, which was suggested to recognize a certain motif around amino acids 30-33 Phe-Ile-Gln-Asp (FIQD). In the present study, analysis of protein sequencing revealed that the cleavage site was between Gln28 and Gly29. The fragment lacking the NH(2)-terminal amino acids 1-28 (tBax(29)) was more apoptotic than wt Bax. The tBax(29)-induced apoptotic cell death was substantially resistant to Bcl-x(L)-mediated rescue, compared with wt Bax, in spite of the complex formation between these two molecules. Together, the tBax(29) would be valuable for the treatment of tumors with high levels of Bcl-x(L) as well as the understanding of Bax-mediated apoptotic processes.


Subject(s)
Apoptosis/drug effects , Calpain/metabolism , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/pharmacology , Adenocarcinoma/pathology , Amino Acid Motifs , Cell Line/drug effects , Codon , Colonic Neoplasms/pathology , Humans , Kidney/embryology , Mutagenesis, Site-Directed , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/pharmacology , Substrate Specificity , Tumor Cells, Cultured/drug effects , bcl-2-Associated X Protein , bcl-X Protein
19.
Immunol Lett ; 81(2): 93-8, 2002 Apr 22.
Article in English | MEDLINE | ID: mdl-11852113

ABSTRACT

CH31 cells have been used for analysis of B cell tolerance, since engagement of membrane immunoglobulin (mIg) results in loss in mitochondrial membrane potential (DeltaPsim), followed by cell death. We have reported that the dominant-negative (dn) form of c-Jun N-terminal kinase (JNK) substantially prevented a loss of DeltaPsim at 24 h, with partial protection around 48 h after anti-IgM stimulation. In this study, we demonstrate that anti-IgM induced a sustained activation of p38 mitogen-activated protein (MAP) kinase. The p38MAP kinase inhibitor SB203580 substantially prevented loss of DeltaPsim at 14 h, with partial prevention (18-24 h) after anti-IgM stimulation. The dnJNK1-mediated prevention of anti-IgM-induced mitochondrial dissipation was enhanced by SB203580 at 42 h, but not 24 h, after stimulation, suggesting that activation of either p38 MAP kinase or JNK may be sufficient for the initiation of early phase of anti-IgM-induced loss of DeltaPsim while both may be necessary in the late phase.


Subject(s)
B-Lymphocytes/physiology , Mitochondria/physiology , Mitogen-Activated Protein Kinases/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , B-Lymphocytes/drug effects , Caspase Inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Immunoglobulin M , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases , Lymphoma, B-Cell , Membrane Potentials , Mitochondria/drug effects , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/immunology , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/pharmacology , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...