Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Mol Ther Oncolytics ; 30: 72-85, 2023 Sep 21.
Article in English | MEDLINE | ID: mdl-37583387

ABSTRACT

A complete resection of tongue cancer is often difficult. We investigate the usefulness of administering G47Δ (teserpaturev), a triple-mutated oncolytic herpes simplex virus type 1, prior to resection. G47Δ exhibits good cytopathic effects and replication capabilities in all head and neck cancer cell lines tested. In an orthotopic SCCVII tongue cancer model of C3H/He mice, an intratumoral inoculation with G47Δ significantly prolongs the survival. Further, mice with orthotopic tongue cancer received neoadjuvant G47Δ (or mock) therapy with or without "hemilateral" resection, the maximum extent avoiding surgical deaths. Neoadjuvant G47Δ and resection led to 10/10 survival (120 days), whereas the survivals for G47Δ alone and resection alone were 6/10 and 5/10, respectively: all control animals died by day 11. Furthermore, 100% survival was achieved with neoadjuvant G47Δ therapy even when the resection area was narrowed to "partial," providing insufficient resection margins, whereas hemilateral resection alone caused death by local recurrence in half of the animals. G47Δ therapy caused increased number of tumor-infiltrating CD8+ and CD4+ cells, increased F4/80+ cells within the residual tongues, and increased expression of immune-related genes in and around the tumor. These results imply that neoadjuvant use of G47Δ is useful for preventing local recurrence after tongue cancer surgery.

2.
Commun Med (Lond) ; 3(1): 40, 2023 Mar 25.
Article in English | MEDLINE | ID: mdl-36966232

ABSTRACT

BACKGROUND: G47∆ is a triple-mutated oncolytic herpes simplex virus type 1 (HSV-1) recently approved as a new drug for malignant glioma in Japan. As the next-generation, we develop armed oncolytic HSV-1 using G47∆ as the backbone. Because oncolytic HSV-1 elicits specific antitumor immunity, interleukin 12 (IL-12) can function as an effective payload to enhance the efficacy. METHODS: We evaluate the optimal methods for expressing IL-12 as a payload for G47∆-based oncolytic HSV-1. Two new armed viruses are generated for evaluation by employing different methods to express IL-12: T-mfIL12 expresses murine IL-12 as a fusion peptide, with the genes of two subunits (p35 and p40) linked by bovine elastin motifs, and T-mIL12-IRES co-expresses the subunits, with the two genes separated by an internal ribosome entry site (IRES) sequence. RESULTS: T-mfIL12 is significantly more efficient in producing IL-12 than T-mIL12-IRES in all cell lines tested, whereas the expression methods do not affect the replication capabilities and cytopathic effects. In two syngeneic mouse subcutaneous tumor models of Neuro2a and TRAMP-C2, T-mfIL12 exhibits a significantly higher efficacy than T-mIL12-IRES when inoculated intratumorally. Furthermore, T-mfIL12 shows a significantly higher intratumoral expression of functional IL-12, causing stronger stimulation of specific antitumor immune responses than T-mIL12-IRES. CONCLUSIONS: The results implicate that a fusion-type expression of IL-12 is a method superior to co-expression of separate subunits, due to higher production of functional IL-12 molecules. This study led to the creation of triple-mutated oncolytic HSV-1 armed with human IL-12 currently used in phase 1/2 trial for malignant melanoma.


Some viruses, including the herpes virus, can be modified so that they can target and kill cancers. These viruses can be loaded with factors that stimulate the immune system, which can help to eradicate cancer cells. Here, we test different methods of loading a cancer-killing version of the herpes virus with interleukin 12, an immune-stimulating factor. We show that one method, which involves loading the virus with the different parts of interleukin 12 fused together, is superior to another, and leads to improved anti-cancer effects in mouse models. These findings have contributed to the creation of a cancer-killing virus that is currently in clinical trials in patients with melanoma.

3.
Mol Ther Oncolytics ; 28: 31-43, 2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36619294

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease known for its dense tumor stroma. Focal adhesion kinase inhibitor (FAKi), a non-receptor type tyrosine kinase inhibitor, reduces the tumor stroma. G47Δ, a third-generation oncolytic herpes simplex virus type 1, destroys tumor cells selectively and induces antitumor immune responses. This study evaluates the efficacy of FAKi and G47Δ in PDAC models in combination with or without immune checkpoint inhibitors. G47Δ was effective in human PDAC cell lines in vitro and in subcutaneous as well as orthotopic tumor models. Transgenic mouse-derived #146 cells were used to generate subcutaneous PDAC tumors with rich stroma in immunocompetent mice. In this #146 tumor model, the efficacy of FAKi was synergistically augmented when combined with G47Δ, which reflected not only a decreased stromal content but also a significant shifting of the tumor microenvironment toward immune stimulation. In transgenic autochthonous PKF mice, a rare model that develops stroma-rich PDAC with a 100% penetrance and resembles human PDAC in various aspects, the prolongation of survival compared with FAKi alone was achieved only when FAKi was combined with G47Δ and immune checkpoint inhibitors. The FAKi combination therapy may be useful to overcome the treatment resistance of stroma-rich PDAC.

4.
Mol Ther Oncolytics ; 23: 402-411, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34853811

ABSTRACT

Treatment options are limited for esophageal carcinoma (EC). G47Δ, a triple-mutated, conditionally replicating herpes simplex virus type 1 (HSV-1), exhibits enhanced killing of tumor cells with high safety features. Here, we studied the efficacy of G47Δ using preclinical models of human EC. In vitro, G47Δ showed efficient cytopathic effects and replication capabilities in all eight human esophageal cancer cell lines tested. In athymic mice harboring subcutaneous tumors of human EC (KYSE180, TE8, and OE19), two intratumoral injections with G47Δ significantly inhibited the tumor growth. To mimic the clinical treatment situations, we established an orthotopic EC model using luciferase-expressing TE8 cells (TE8-luc). An intratumoral injection with G47Δ markedly inhibited the growth of orthotopic TE8-luc tumors in athymic mice. Furthermore, we evaluated the safety of applying G47Δ to the esophagus in mice. A/J mice inoculated intraesophageally or administered orally with G47Δ (107 plaque-forming units [pfu]) survived for more than 2 months without remarkable symptoms, whereas the majority with wild-type HSV-1 (106 pfu) deteriorated within 10 days. PCR analyses showed that the G47Δ DNA was confined to the esophagus after intraesophageal inoculation and was not detected in major organs after oral administration. Our results provide a rationale for the clinical use of G47Δ for treating EC.

5.
Mol Ther Oncolytics ; 22: 388-398, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34553027

ABSTRACT

The prognosis of oral squamous cell carcinoma (OSCC) largely depends on the control of lymph node metastases. We evaluate the therapeutic efficacy of G47Δ, a third-generation oncolytic herpes simplex virus type 1 (HSV-1), in mouse tongue cancer models. Intratumoral injection with G47Δ prolonged the survival in all orthotopic models investigated. In both athymic and immunocompetent models, G47Δ injected into the tongue cancer swiftly traffics to the draining cervical lymph nodes and suppresses lymph node metastases. In the immunocompetent KLN205-MUC1 model, in which the metastatic cascade that tongue cancer patients commonly experience is reproduced, intratumoral G47Δ injection even immediately prior to a tumor resection prolonged survival. Cervical lymph nodes 18 h after G47Δ treatment showed the presence of G47Δ infection and an increase in CD69-positive cells, indicating an immediate activation of T cells. Furthermore, G47Δ injected directly into enlarged metastatic lymph nodes significantly prolonged the survival at an advanced stage. Whereas intratumorally injected oncolytic HSV-1 does not readily circulate in the blood stream, G47Δ is shown to traffic in the lymphatics swiftly. The use of G47Δ can lead to entirely new treatment strategies for tongue cancer and other OSCC at all clinical stages.

6.
Mol Ther Oncolytics ; 22: 129-142, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34514094

ABSTRACT

Oncolytic virus therapy can increase the immunogenicity of tumors and remodel the immunosuppressive tumor microenvironment, leading to an increased antitumor response to immune-checkpoint inhibitors. Here, we investigated the therapeutic potential of G47Δ, a third-generation oncolytic herpes simplex virus type 1, in combination with immune-checkpoint inhibitors using various syngeneic murine subcutaneous tumor models. Intratumoral inoculations with G47Δ and systemic anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibody administration caused an enhanced antitumor activity when combined and worked synergistically. Conversely, the efficacy of G47Δ in combination with anti-programmed cell death protein-1 (PD-1) antibody was equivalent to that of the anti-PD-1 antibody alone in all murine models examined. The combination of intratumoral G47Δ and systemic anti-CTLA-4 antibody was shown to recruit effector T cells into the tumor efficiently while decreasing regulatory T cells. Furthermore, a wide range of gene signatures related to inflammation, lymphoid lineage, and T cell activation was highly upregulated with the combination therapy, suggesting the conversion of immune-insusceptible tumors to immune susceptible. The therapeutic effect proved tumor specific and long lasting. Immune cell subset depletion studies demonstrated that CD4+ T cells were required for synergistic curative activity. The results depict the dynamics of immune modulation of the tumor microenvironment and provide a clinical rationale for using G47Δ with immune checkpoint inhibitors.

7.
Biol Pharm Bull ; 44(3): 293-297, 2021.
Article in English | MEDLINE | ID: mdl-33642541

ABSTRACT

Chemotherapy regimen management is one of the most important oncology pharmacy practices, because chemotherapy is conducted according to the registered regimens. In this study, we evaluated the pharmaceutical practice that assumes the initial confirmation of chemotherapy regimens and the quality of practice sharing between oncology-specialized and non-specialized pharmacists in regimen management committee. Pharmacists initially confirmed the applied regimen prescribed by physicians regarding chemotherapeutic agents and prophylactic supportive care medicines. Following confirmation, the regimens were reviewed by the Hokkaido University Hospital Regimen Management Committee. A total of 233 regimens were reviewed by the committee over three years. In total, 110 pharmaceutical inquiries were conducted, 45% of inquiries were concerning chemotherapeutic agents, of which approximately half were regarding supportive care medicines. Most inquiries were regarding premedication, followed by those on administration time, solvent of infusion medicines, and dosage. Correction was performed for 84.5% of inquiries. There was no significant difference in inquiry rates between practice and trial regimens. We have entrusted the first basic regimen review according to the checklist, creation of the chemotherapy plan document, and registry of the adopted regimens in the ordering system from oncology-certified pharmacists to non-certified pharmacists. Basic regimen review was well conducted by a non-certified pharmacist, and a more advanced review was additionally performed by certified pharmacists. In conclusion, we demonstrated the utility of pharmaceutical confirmation in a chemotherapeutic regimen review, suitable review coverage, and quality practice sharing between oncology-certified and non-certified pharmacists, which is one of the recommended methods in chemotherapy regimen review.


Subject(s)
Antineoplastic Agents/therapeutic use , Pharmacists/organization & administration , Pharmacy Service, Hospital/organization & administration , Practice Patterns, Pharmacists' , Drug Utilization Review , Hospitals, University/organization & administration , Humans , Japan
8.
Mol Ther Oncolytics ; 18: 535-545, 2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32995479

ABSTRACT

G47Δ is a triple-mutated oncolytic herpes simplex virus type 1 designed to induce antitumor immune responses efficiently. We examine the usefulness of G47Δ as a neoadjuvant therapy for radiofrequency ablation (RFA), a standard local treatment for certain cancers such as liver cancer, but remote recurrences within the same organ often occur. In A/J mice harboring bilateral subcutaneous Neuro2a tumors, the left tumors were treated with G47Δ intratumoral injections followed by RFA. Whereas the RFA-treated tumors were all eradicated, the growth of the right tumors was evaluated and tumor-infiltrating lymphocytes were analyzed. The G47Δ+RFA treatment caused smaller volumes of right tumors, accompanied by increased CD8+/CD45+ T cells, compared with G47Δ monotherapy. After depletion of CD8+ T cells, the enhanced efficacy on the contralateral tumors was completely abolished. Neoadjuvant G47Δ led to rejection of rechallenged tumors, which was caused by efficient induction of specific antitumor immune responses shown by enzyme-linked immunospot (ELISPOT) assays. Treatment of tumor-harboring animals with an anti-programmed cell death 1 ligand 1 (PD-L1) antibody led to even greater efficacy on contralateral tumors. Our study indicates that the neoadjuvant use of G47Δ effectively enhances the efficacy of RFA via CD8+ T cell-dependent immunity that is further augmented by an immune checkpoint inhibitor.

9.
Mol Ther Oncolytics ; 17: 205-215, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32346610

ABSTRACT

Advanced gastric cancer, especially scirrhous gastric cancer with peritoneal dissemination, remains refractory to conventional therapies. G47Δ, a third-generation oncolytic herpes simplex virus type 1, is an attractive novel therapeutic agent for solid cancer. In this study, we investigated the therapeutic potential of G47Δ for human gastric cancer. In vitro, G47Δ showed good cytopathic effects and replication capabilities in nine human gastric cancer cell lines tested. In vivo, intratumoral inoculations with G47Δ (2 × 105 or 1 × 106 plaque-forming units [PFU]) significantly inhibited the growth of subcutaneous tumors (MKN45, MKN74, and 44As3). To evaluate the efficacy of G47Δ for advanced-stage models of gastric cancer, we generated an orthotopic tumor model and peritoneal dissemination models of human scirrhous gastric cancer (MKN45-luc and 44As3Luc), which have features mimicking intractable scirrhous cancer patients. G47Δ (1 × 106 PFU) was constantly efficacious whether administered intratumorally or intraperitoneally in the clinically relevant models. Notably, G47Δ injected intraperitoneally readily distributed to, and selectively replicated in, disseminated tumors. Furthermore, flow cytometric analyses of tumor-infiltrating cells in subcutaneous tumors revealed that intratumoral G47Δ injections markedly decreased M2 macrophages while increasing M1 macrophages and natural killer (NK) cells. These findings indicate the usefulness of G47Δ for treating human gastric cancer, including scirrhous gastric cancer and the ones in advanced stages.

10.
Cancer Sci ; 103(6): 1051-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22429880

ABSTRACT

CADM1, a member of the immunoglobulin superfamily cell adhesion molecule, acts as a tumor suppressor in a variety of human cancers. CADM1 is also ectopically expressed in adult T-cell leukemia (ATL), conferring an invasive phenotype characteristic to ATL. Therefore, CADM1 plays dual roles in human oncogenesis. Here, we investigate the roles of CADM1 in small cell lung cancer (SCLC). Immunohistochemistry demonstrates that 10 of 35 (29%) primary SCLC tumors express CADM1 protein. Western blotting and RT-PCR analyses reveal that CADM1 is significantly expressed in 11 of 14 SCLC cells growing in suspension cultures but in neither of 2 SCLC cells showing attached growth to plastic dishes, suggesting that CADM1 is involved in anchorage-independent growth in SCLC. In the present study, we demonstrate that SCLC expresses a unique splicing variant of CADM1 (variant 8/9) containing additional extracellular fragments corresponding to exon 9 in addition to variant 8, a common isoform in epithelia. Variant 8/9 of CADM1 is almost exclusively observed in SCLC and testis, although this variant protein localizes along the membrane and shows similar cell aggregation activity to variant 8. Interestingly, both variant 8/9 and variant 8 of CADM1 show enhanced tumorigenicity in nude mice when transfected into SBC5, a SCLC cell lacking CADM1. Inversely, suppression of CADM1 expression by shRNA reduced spheroid-like cell aggregation of NCI-H69, an SCLC cell expressing a high amount of CADM1. These findings suggest that CADM1 enhances the malignant features of SCLC, as is observed in ATL, and could provide a molecular marker specific to SCLC.


Subject(s)
Cell Adhesion Molecules/metabolism , Immunoglobulins/metabolism , Small Cell Lung Carcinoma/metabolism , Animals , Cell Adhesion , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/genetics , Humans , Immunoglobulins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Tumor Cells, Cultured , Tumor Suppressor Proteins/metabolism
11.
Breast Cancer ; 19(3): 242-52, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21526423

ABSTRACT

BACKGROUND: The tumor suppressor genes CADM1/TSLC1 and DAL-1/4.1B are frequently inactivated by promoter methylation in non-small cell lung cancer. The proteins they encode, CADM1 and 4.1B, form a complex in human epithelial cells and are involved in cell-cell adhesion. METHODS: Expression of CADM1 and 4.1B proteins was examined by immunohistochemistry in 67 primary breast cancer and adjacent noncancerous tissues. CADM1 and 4.1B messenger RNA (mRNA) was detected by reverse-transcription polymerase chain reaction (RT-PCR). The methylation status of the CADM1 and 4.1B promoters was determined quantitatively by bisulfite treatment followed by pyrosequencing. RESULTS: CADM1 and 4.1B protein signals were detected along the cell membrane in normal mammary epithelia. By contrast, 47 (70%) and 49 (73%) of 67 primary breast cancers showed aberrant CADM1 and 4.1B staining, respectively. Aberrant CADM1 staining was more frequently observed in pT2 and pT3 tumors and for stages II and III (P = 0.045 and P = 0.020, respectively), while aberrant 4.1B staining was more often observed in tumors with lymph node metastasis, for pT2 and pT3 tumors, and for stages II and III (P = 0.0058, P = 0.0098, and P = 0.0007, respectively). Furthermore, aberrant CADM1 and 4.1B expression was preferentially observed in invasive relative to noninvasive lesions from the same specimen (P = 0.036 and P = 0.0009, respectively). Finally, hypermethylation of CADM1 and 4.1B genes was detected in 46% and 42% of primary breast cancers, respectively. CONCLUSIONS: Our findings suggest that aberrant CADM1 and 4.1B expression is involved in progression of breast cancer, especially in invasion into the stroma and metastasis.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion Molecules/genetics , Immunoglobulins/genetics , Membrane Proteins/genetics , Adult , Aged , Aged, 80 and over , Breast Neoplasms/metabolism , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , DNA Methylation , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoglobulins/metabolism , Membrane Proteins/metabolism , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Promoter Regions, Genetic , Reference Values , Stromal Cells/pathology , Tumor Suppressor Proteins/genetics
12.
Proc Natl Acad Sci U S A ; 108(37): 15486-91, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21876165

ABSTRACT

The inositol 1,4,5-trisphosphate (IP(3)) receptor (IP(3)R) is an intracellular Ca(2+) release channel, and its opening is controlled by IP(3) and Ca(2+). A single IP(3) binding site and multiple Ca(2+) binding sites exist on single subunits, but the precise nature of the interplay between these two ligands in regulating biphasic dependence of channel activity on cytosolic Ca(2+) is unknown. In this study, we visualized conformational changes in IP(3)R evoked by various concentrations of ligands by using the FRET between two fluorescent proteins fused to the N terminus of individual subunits. IP(3) and Ca(2+) have opposite effects on the FRET signal change, but the combined effect of these ligands is not a simple summative response. The bell-shaped Ca(2+) dependence of FRET efficiency was observed after the subtraction of the component corresponding to the FRET change evoked by Ca(2+) alone from the FRET changes evoked by both ligands together. A mutant IP(3)R containing a single amino acid substitution at K508, which is critical for IP(3) binding, did not exhibit this bell-shaped Ca(2+) dependence of the subtracted FRET efficiency. Mutation at E2100, which is known as a Ca(2+) sensor, resulted in ∼10-fold reduction in the Ca(2+) dependence of the subtracted signal. These results suggest that the subtracted FRET signal reflects IP(3)R activity. We propose a five-state model, which implements a dual-ligand competition response without complex allosteric regulation of Ca(2+) binding affinity, as the mechanism underlying the IP(3)-dependent regulation of the bell-shaped relationship between the IP(3)R activity and cytosolic Ca(2+).


Subject(s)
Calcium/metabolism , Cytosol/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Ion Channel Gating , Animals , Bacterial Proteins/metabolism , Calcium/pharmacology , Cytosol/drug effects , Fluorescence Resonance Energy Transfer , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Ion Channel Gating/drug effects , Ligands , Luminescent Proteins/metabolism , Mice , Models, Biological , Recombinant Fusion Proteins/metabolism
13.
Genes Cells ; 16(7): 791-802, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21672091

ABSTRACT

CADM1 is a multifunctional cell adhesion molecule expressed predominantly in the nerve system, testis and lung. The expression of the Cadm1 gene is induced during the neural differentiation of murine embryonal carcinoma P19 cells by treatment with retinoic acid (RA). Here, we show that the suppression of CADM1 expression using RNAi interfered with P19 cell aggregation and reduced cell populations expressing MAP2 after RA treatment. Nonaggregated P19 cells were not differentiated into neurons, suggesting that CADM1 participates in the aggregate formation and neuronal differentiation of P19 in vitro. A luciferase assay of a series of deletion mutants of the CADM1 promoter localized an RA-responsive cis-acting element to an approximately 90-bp fragment upstream of the translational start site. This element contains a putative binding site for transcription factor Sp1, named Sp1-binding site-1 (Sp1BS-1). Sp1BS-1 and adjacent Sp1-binding sites (Sp1BS-2 and Sp1BS-3) showed enhanced transcriptional activity by RA. Moreover, a chromatin immunoprecipitation showed that RA receptor (RAR)α was associated with a DNA fragment containing Sp1BS-1, whereas suppression of RARα expression using siRNA reduced the responsiveness of the CADM1 promoter to RA. These results suggest that Sp1 plays a critical role in RA-induced CADM1 expression through possible interaction with RARα in the neural differentiation of P19.


Subject(s)
Carcinoma, Embryonal/pathology , Cell Adhesion Molecules/genetics , Cell Differentiation/genetics , Immunoglobulins/genetics , Neural Stem Cells/drug effects , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Animals , Binding Sites , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/metabolism , Immunoglobulins/metabolism , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sp1 Transcription Factor/metabolism , Tumor Cells, Cultured
14.
J Biol Chem ; 283(31): 21579-87, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18505727

ABSTRACT

ATP enhances Ca(2+) release from inositol (1,4,5)-trisphosphate receptors (InsP(3)R). However, the three isoforms of InsP(3)R are reported to respond to ATP with differing sensitivities. Ca(2+) release through InsP(3)R1 is positively regulated at lower ATP concentrations than InsP(3)R3, and InsP(3)R2 has been reported to be insensitive to ATP modulation. We have reexamined these differences by studying the effects of ATP on InsP(3)R2 and InsP(3)R3 expressed in isolation on a null background in DT40 InsP(3)R knockout cells. We report that the Ca(2+)-releasing activity as well as the single channel open probability of InsP(3)R2 was enhanced by ATP, but only at submaximal InsP(3) levels. Further, InsP(3)R2 was more sensitive to ATP modulation than InsP(3)R3 under similar experimental conditions. Mutations in the ATPB sites of InsP(3)R2 and InsP(3)R3 were generated, and the functional consequences of these mutations were tested. Surprisingly, mutation of the ATPB site in InsP(3)R3 had no effect on ATP modulation, suggesting an additional locus for the effects of ATP on this isoform. In contrast, ablation of the ATPB site of InsP(3)R2 eliminated the enhancing effects of ATP. Furthermore, this mutation had profound effects on the patterns of intracellular calcium signals, providing evidence for the physiological significance of ATP binding to InsP(3)R2.


Subject(s)
Adenosine Triphosphate/chemistry , Calcium/chemistry , Inositol 1,4,5-Trisphosphate Receptors/physiology , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Calcium/metabolism , Calcium Signaling , Cell Line , Chickens , DNA Mutational Analysis , Glutathione Transferase/metabolism , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Models, Biological , Mutation , Probability , Signal Transduction , Transfection
15.
Mod Rheumatol ; 17(2): 137-43, 2007.
Article in English | MEDLINE | ID: mdl-17437169

ABSTRACT

IP(3)R2 and IP(3)R3 double knock-out mice present with exocrine dysfunctions such as secretion deficits of saliva and pancreatic juice. Therefore, we investigated whether the presence of antibodies to IP(3)Rs could be found in patients with Sjögren's syndrome, and the location of the epitopes. Subjects included 35 primary Sjögren's syndrome, 39 secondary Sjögren's syndrome, 144 rheumatoid arthritis, and 96 other connective tissue disease patients. As controls, 33 healthy subjects were included. Immunoblot was conducted using recombinant proteins IP(3)R1, IP(3)R2, and IP(3)R3 made from full-length cDNA, and core, T604, and EL for epitope mapping. Antibodies to IP(3)R1 in sera from patients with primary Sjögren's syndrome, secondary Sjögren's syndrome, and rheumatoid arthritis were found to be positive in 17 of 35 (48.6%), 13 of 39 (33%), and 34 of 124 (27.4%) cases, respectively. These frequencies were significantly higher than the 1 of 33 (3.0%) found in normal healthy subjects. The frequency of anti-IP(3)R2 antibodies in rheumatoid arthritis was found to be higher than those found in Sjögren's syndrome, systemic lupus erythematosus, and systemic sclerosis. Anti-IP(3)R2 antibodies found in rheumatoid arthritis primarily recognized residues 578-2171 of the internal coupling and regulatory domain. On the other hand, anti-IP(3)R1 found in Sjögren's syndrome recognized residues 224-604 of the core protein IP(3)R1. Anti-IP(3)R1 antibodies were present in 48.6% of primary Sjögren's syndrome and in 3.0% of normal healthy subjects. Anti-IP(3)R2 antibodies were detected most frequently in rheumatoid arthritis. Locations of the antigenic epitopes were found to differ among the disease conditions.


Subject(s)
Autoantibodies/blood , Inositol 1,4,5-Trisphosphate Receptors/immunology , Rheumatic Diseases/immunology , Sjogren's Syndrome/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Antinuclear/blood , Female , Humans , Male , Middle Aged , Reference Values
16.
J Biol Chem ; 282(17): 12755-64, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17327232

ABSTRACT

Three isoforms of the inositol 1,4,5-trisphosphate (IP(3)) receptor (IP(3)R), IP(3)R1, IP(3)R2, and IP(3)R3, have different IP(3)-binding affinities and cooperativities. Here we report that the amino-terminal 604 residues of three mouse IP(3)R types exhibited K(d) values of 49.5 +/- 10.5, 14.0 +/- 3.5, and 163.0 +/- 44.4 nm, which are close to the intrinsic IP(3)-binding affinity previously estimated from the analysis of full-length IP(3)Rs. In contrast, residues 224-604 of IP(3)R1 and IP(3)R2 and residues 225-604 of IP(3)R3, which contain the IP(3)-binding core domain but not the suppressor domain, displayed an almost identical IP(3)-binding affinity with a K(d) value of approximately 2 nm. Addition of 100-fold excess of the suppressor domain did not alter the IP(3)-binding affinity of the IP(3)-binding core domain. Artificial chimeric proteins in which the suppressor domain was fused to the IP(3)-binding core domain from different isoforms exhibited IP(3)-binding affinity significantly different from those of the proteins composed of the native combination of the suppressor domain and the IP(3)-binding core domain. Systematic mutagenesis analyses showed that amino acid residues critical for type-3 receptor-specific IP(3)-binding affinity are involved in Glu-39, Ala-41, Asp-46, Met-127, Ala-154, Thr-155, Leu-162, Trp-168, Asn-173, Asn-176, and Val-179. These results indicate that the IP(3)-binding affinity of IP(3)Rs is specifically tuned through the intramolecular attenuation of IP(3)-binding affinity of the IP(3)-binding core domain by the amino-terminal suppressor domain. Moreover, the functional diversity in ligand sensitivity among IP(3)R isoforms originates from at least the structural difference identified on the suppressor domain.


Subject(s)
Amino Acid Substitution , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Inositol 1,4,5-Trisphosphate/chemistry , Animals , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Kinetics , Ligands , Mice , Protein Binding/genetics , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary/genetics , Structure-Activity Relationship
17.
Clin Rheumatol ; 26(9): 1571-4, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17131037

ABSTRACT

In this report, we present a 63-year-old woman who had limited cutaneous systemic sclerosis and subsequently developed typical primary biliary cirrhosis after an acute myocardial infarction. The patient initially developed Raynaud's phenomenon, and 4 years later visited the clinic in 1994 complaining of abdominal distress, xerostomia, and xerophthalmia. A diagnosis of limited cutaneous systemic sclerosis was based on Raynaud's phenomenon, sclerodactyly and anti-centromere antibodies. She was also found to have anti-inositol 1,4,5-trisphosphate receptor 3 (IP(3)R3) antibodies, but anti-mitochondrial antibodies were only weakly positive. Seven years later, she developed vertigo and nausea, and was hospitalized due to complaints of an oppressive sensation of the anterior chest. Electrocardiogram results showed a reduction of R waves and ST segment elevation in II, III, and aVf leads. Coronary angiography showed 99% obstruction of the left anterior descending artery and 50% of stenosis of the right coronary artery. Three years later, the patient was noted to have anti-mitochondrial antibodies. Retrospective analysis of the patient's sera showed that IP(3)R3 antibodies were decreasing. Since myocardium is particularly rich in mitochondria, it is thought that myocardial infarction may have been the triggering event that initiated antigen-presenting cells to selectively induce an anti-mitochondrial antibody response.


Subject(s)
Autoantibodies/immunology , Liver Cirrhosis, Biliary/immunology , Mitochondria/immunology , Myocardial Infarction/complications , Scleroderma, Limited/immunology , Female , HMGB1 Protein/immunology , Humans , Inositol 1,4,5-Trisphosphate Receptors/immunology , Liver Cirrhosis, Biliary/complications , Middle Aged , Myocardial Infarction/immunology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/immunology , Scleroderma, Limited/complications
18.
J Biol Chem ; 280(11): 10305-17, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15632133

ABSTRACT

We isolated cDNAs encoding type 2 and type 3 inositol 1,4,5-trisphosphate (IP(3)) receptors (IP(3)R2 and IP(3)R3, respectively) from mouse lung and found a novel alternative splicing segment, SI(m2), at 176-208 of IP(3)R2. The long form (IP(3)R2 SI(m2)(+)) was dominant, but the short form (IP(3)R2 SI(m2)(-)) was detected in all tissues examined. IP(3)R2 SI(m2)(-) has neither IP(3) binding activity nor Ca(2+) releasing activity. In addition to its reticular distribution, IP(3)R2 SI(m2)(+) is present in the form of clusters in the endoplasmic reticulum of resting COS-7 cells, and after ATP or Ca(2+) ionophore stimulation, most of the IP(3)R2 SI(m2)(+) is in clusters. IP(3)R3 is localized uniformly on the endoplasmic reticulum of resting cells and forms clusters after ATP or Ca(2+) ionophore stimulation. IP(3)R2 SI(m2)(-) does not form clusters in either resting or stimulated cells. IP(3) binding-deficient site-directed mutants of IP(3)R2 SI(m2)(+) and IP(3)R3 fail to form clusters, indicating that IP(3) binding is involved in the cluster formation by these isoforms. Coexpression of IP(3)R2 SI(m2)(-) prevents stimulus-induced IP(3)R clustering, suggesting that IP(3)R2 SI(m2)(-) functions as a negative coordinator of stimulus-induced IP(3)R clustering. Expression of IP(3)R2 SI(m2)(-) in CHO-K1 cells significantly reduced ATP-induced Ca(2+) entry, but not Ca(2+) release, suggesting that the novel splice variant of IP(3)R2 specifically influences the dynamics of the sustained phase of Ca(2+) signals.


Subject(s)
Alternative Splicing , Calcium Channels/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Adenosine Triphosphate/chemistry , Amino Acid Sequence , Animals , Blotting, Western , CHO Cells , COS Cells , Calcium/metabolism , Calcium Channels/chemistry , Cell Line , Cloning, Molecular , Cricetinae , Cytoplasm/metabolism , DNA, Complementary/metabolism , Endoplasmic Reticulum/metabolism , Green Fluorescent Proteins/metabolism , Immunoprecipitation , Inositol 1,4,5-Trisphosphate Receptors , Insecta , Ionophores/pharmacology , Kinetics , Lung/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Microsomes/metabolism , Molecular Sequence Data , Multigene Family , Mutagenesis, Site-Directed , Rats , Receptors, Cytoplasmic and Nuclear/chemistry , Recombinant Proteins/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Time Factors , Tissue Distribution
19.
J Biol Chem ; 280(8): 6816-22, 2005 Feb 25.
Article in English | MEDLINE | ID: mdl-15583010

ABSTRACT

The inositol 1,4,5-trisphosphate (IP(3)) receptor (IP(3)R) Ca(2+) channel plays pivotal roles in many aspects of physiological and pathological events. It was previously reported that IP(3)R forms clusters on the endoplasmic reticulum when cytosolic Ca(2+) concentration ([Ca(2+)](C)) is elevated. However, the molecular mechanism of IP(3)R clustering remains largely unknown, and thus its physiological significance is far from clear. In this study we found that the time course of clustering of green fluorescent protein-tagged IP(3)R type 1 (GFP-IP(3)R1), evoked by IP(3)-generating agonists, did not correlate with [Ca(2+)](C) but seemed compatible with cytoplasmic IP(3) concentration. IP(3) production alone induced GFP-IP(3)R1 clustering in the absence of a significant increase in [Ca(2+)](C) but elevated [Ca(2+)](C) without IP(3) production did not. Moreover IP(3)R1 mutants that do not undergo an IP(3)-induced conformational change failed to form clusters. Thus, IP(3)R clustering is induced by its IP(3)-induced conformational change to the open state. We also found that GFP-IP(3)R1 clusters colocalized with ERp44, a luminal protein of endoplasmic reticulum that inhibits its channel activity. This is the first example of ligand-induced clustering of a ligand-gated channel protein.


Subject(s)
Calcium Channels/physiology , Receptor Aggregation , Receptors, Cytoplasmic and Nuclear/physiology , Animals , COS Cells , Calcium/analysis , Calcium Channels/chemistry , Calcium Channels/genetics , Chlorocebus aethiops , Cytosol/chemistry , Green Fluorescent Proteins , Inositol 1,4,5-Trisphosphate/analysis , Inositol 1,4,5-Trisphosphate Receptors , Mice , Microscopy, Fluorescence , Protein Conformation , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Transfection
20.
Biochem J ; 377(Pt 2): 299-307, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-12968951

ABSTRACT

The type 1 inositol 1,4,5-trisphosphate receptor (IP(3)R1) is an intracellular Ca(2+) channel protein that plays crucial roles in generating complex Ca(2+) signalling patterns. IP(3)R1 consists of three domains: a ligand-binding domain, a regulatory domain and a channel domain. In order to investigate the function of these domains in its gating machinery and the physiological significance of specific cleavage by caspase 3 that is observed in cells undergoing apoptosis, we utilized various IP(3)R1 constructs tagged with green fluorescent protein (GFP). Expression of GFP-tagged full-length IP(3)R1 or IP(3)R1 lacking the ligand-binding domain in HeLa and COS-7 cells had little effect on cells' responsiveness to an IP(3)-generating agonist ATP and Ca(2+) leak induced by thapsigargin. On the other hand, in cells expressing the caspase-3-cleaved form (GFP-IP(3)R1-casp) or the channel domain alone (GFP-IP(3)R1-ES), both ATP and thapsigargin failed to induce increase of cytosolic Ca(2+) concentration. Interestingly, store-operated (-like) Ca(2+) entry was normally observed in these cells, irrespective of thapsigargin pre-treatment. These findings indicate that the Ca(2+) stores of cells expressing GFP-IP(3)R1-casp or GFP-IP(3)R1-ES are nearly empty in the resting state and that these proteins continuously leak Ca(2+). We therefore propose that the channel domain of IP(3)R1 tends to remain open and that the large regulatory domain of IP(3)R1 is necessary to keep the channel domain closed. Thus cleavage of IP(3)R1 by caspase 3 may contribute to the increased cytosolic Ca(2+) concentration often observed in cells undergoing apoptosis. Finally, GFP-IP(3)R1-casp or GFP-IP(3)R1-ES can be used as a novel tool to deplete intracellular Ca(2+) stores.


Subject(s)
Calcium Channels/chemistry , Calcium Channels/metabolism , Calcium/metabolism , Caspases/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Apoptosis , COS Cells , Calcium Channels/genetics , Caspase 3 , Cell Line , Green Fluorescent Proteins , HeLa Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors , Ion Channel Gating , Ion Transport , Luminescent Proteins/genetics , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Fusion Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...