Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 28(12): 2633-2645, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35381070

ABSTRACT

PURPOSE: Osteosarcoma, the most common bone malignancy in children, has a poor prognosis, especially when the tumor metastasizes to the lungs. Therefore, novel therapeutic strategies targeting both proliferation and metastasis of osteosarcoma are required. Podoplanin (PDPN) is expressed by various tumors and is associated with tumor-induced platelet activation via its interaction with C-type lectin-like receptor 2 (CLEC-2) on platelets. We previously found that PDPN contributed to osteosarcoma growth and metastasis through platelet activation; thus, in this study, we developed an anti-PDPN humanized antibody and evaluated its effect on osteosarcoma growth and metastasis. EXPERIMENTAL DESIGN: Nine osteosarcoma cell lines and two osteosarcoma patient-derived cells were collected, and we evaluated the efficacy of the anti-DPN-neutralizing antibody PG4D2 and the humanized anti-PDPN antibody AP201, which had IgG4 framework region. The antitumor and antimetastasis effect of PG4D2 and AP201 were examined in vitro and in vivo. In addition, growth signaling by the interaction between PDPN and CLEC-2 was analyzed using phospho-RTK (receptor tyrosine kinase) array, growth assay, or immunoblot analysis under the supression of RTKs by knockout and inhibitor treatment. RESULTS: We observed that PG4D2 treatment significantly suppressed tumor growth and pulmonary metastasis in osteosarcoma xenograft models highly expressing PDPN. The contribution of PDGFR activation by activated platelet releasates to osteosarcoma cell proliferation was confirmed, and the humanized antibody, AP201, suppressed in vivo osteosarcoma growth and metastasis without significant adverse events. CONCLUSIONS: Targeting PDPN with a neutralizing antibody against PDPN-CLEC-2 without antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity is a novel therapeutic strategy for PDPN-positive osteosarcoma.


Subject(s)
Bone Neoplasms , Lectins, C-Type , Lung Neoplasms , Membrane Glycoproteins , Osteosarcoma , Antibodies, Neutralizing , Bone Neoplasms/drug therapy , Cell Line, Tumor , Humans , Lung Neoplasms/metabolism , Osteosarcoma/drug therapy
2.
Hum Mol Genet ; 31(21): 3652-3671, 2022 10 28.
Article in English | MEDLINE | ID: mdl-35388883

ABSTRACT

Wilson's disease (WD) is a copper metabolic disorder caused by a defective ATP7B function. Conventional therapies cause severe side effects and significant variation in efficacy, according to cohort studies. Thus, exploring new therapeutic approaches to prevent progression to liver failure is urgent. To study the physiology and pathology of WD, immortalized cell lines and rodent WD models have been used conventionally; however, a large gap remains among different species as well as in genetic backgrounds among individuals. We generated induced pluripotent stem cells (iPSCs) from four WD patients carrying compound heterozygous mutations in the ATP7B gene. ATP7B loss- and gain-of-functions were further manifested with ATP7B-deficient iPSCs and heterozygously corrected R778L WD patient-derived iPSCs using CRISPR-Cas9-based gene editing. Although the expression of ATP7B protein varied among WD-specific hepatocytes differentiated from these iPSCs, the expression and secretion of ceruloplasmin (Cp), a downstream copper carrier in plasma, were consistently decreased in WD patient-derived and ATP7B-deficient hepatocytes. A transcriptome analysis detected abnormalities in the retinoid signaling pathway and lipid metabolism in WD-specific hepatocytes. Drug screening using WD patient-derived hepatocytes identified retinoids as promising candidates for rescuing Cp secretion. All-trans retinoic acid also alleviates reactive oxygen species production induced by lipid accumulation in WD-specific hepatocytes treated with oleic acid. These patient-derived iPSC-based hepatic models function as effective platforms for the development of potential therapeutics for hepatic steatosis in WD and other fatty liver diseases.


Subject(s)
Hepatolenticular Degeneration , Humans , Hepatolenticular Degeneration/drug therapy , Hepatolenticular Degeneration/genetics , Ceruloplasmin/genetics , Ceruloplasmin/metabolism , Copper/metabolism , Retinoids/metabolism , Retinoids/therapeutic use , Copper-Transporting ATPases/genetics , Hepatocytes/metabolism , Oxidative Stress , Mutation
3.
Stem Cell Res ; 61: 102744, 2022 05.
Article in English | MEDLINE | ID: mdl-35292424

ABSTRACT

DiGeorge syndrome (22q11.2 deletion syndrome, or CATCH22 syndrome), caused by hemizygous deletion of chromosome 22q11.2, results in the poor development of multiple organs. Here we have generated DiGeorge syndrome-specific human induced pluripotsnt stem cells (hiPSCs) derived from four patients. These established hiPSC lines showed self-renewal and pluripotency and carried a hemizygous deletion in 22q11.2. Since the molecular pathogenesis of DiGeorge syndrome caused by the 22q11.2 deletion is largely unknown, these cell resources will be useful for recapitulating disease phenotypes and for developing new therapies for DiGeorge syndrome.


Subject(s)
DiGeorge Syndrome , Induced Pluripotent Stem Cells , Chromosomes, Human, Pair 2 , DiGeorge Syndrome/genetics , Humans , Phenotype
4.
iScience ; 25(1): 103525, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35106457

ABSTRACT

Non-genetically modified somatic cells can only be inefficiently and stochastically reprogrammed to pluripotency by exogenous expression of reprogramming factors. Low competence of natural reprogramming factors may prevent the majority of cells to successfully and synchronously reprogram. Here we screened DNA-interacting amino acid residues in the zinc-finger domain of KLF4 for enhanced reprogramming efficiency using alanine-substitution scanning methods. Identified KLF4 L507A mutant accelerated and stabilized reprogramming to pluripotency in both mouse and human somatic cells. By testing all the variants of L507 position, variants with smaller amino acid residues in the KLF4 L507 position showed higher reprogramming efficiency. L507A bound more to promoters or enhancers of pluripotency genes, such as KLF5, and drove gene expression of these genes during reprogramming. Molecular dynamics simulations predicted that L507A formed additional interactions with DNA. Our study demonstrates how modifications in amino acid residues of DNA-binding domains enable next-generation reprogramming technology with engineered reprogramming factors.

5.
Stem Cell Res ; 53: 102363, 2021 05.
Article in English | MEDLINE | ID: mdl-34087992

ABSTRACT

ISL1 encodes a member of the LIM/homeodomain family of transcription factors. This encoded protein plays central roles in the development of motor neuron, pancreas, and secondary heart field. Here we generated heterozygous fluorescent reporters of the ISL1 gene in human induced pluripotent stem cells (hiPSCs). CRISPR/Cas9 genome editing technology was employed to knock-in 2A-tdTomato and EF1 alpha promoter-driven Bleomycin resistance gene to the translational ISL1 C-terminal region. The resulting ISL1-TEZ lines showed tdTomato fluorescence upon motor neuron differentiation. These reporter iPSC lines provide opportunity for monitoring and purifying these related cell lineages.


Subject(s)
Gene Editing , Induced Pluripotent Stem Cells , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Humans , Luminescent Proteins , Red Fluorescent Protein
6.
Stem Cell Res ; 53: 102337, 2021 05.
Article in English | MEDLINE | ID: mdl-33901816

ABSTRACT

Adrenoleukodystrophy (ALD) is an X-linked genetic disorder, characterized by demyelination in the central nervous system and adrenal insufficiency. Human induced pluripotent stem cell (hiPSC) lines derived from two Japanese male patients with ALD were generated from skin fibroblasts using retroviral vectors. The generated hiPSC lines showed self-renewal and pluripotency, and carried either a missense or a nonsense mutation in ABCD1 gene. Since the molecular pathogenesis caused by ABCD1 dysfunction remains unclear, these cell resources provide useful tools to establish disease models and to develop new therapies for X-ALD.


Subject(s)
Adrenoleukodystrophy , Genetic Diseases, X-Linked , Induced Pluripotent Stem Cells , ATP Binding Cassette Transporter, Subfamily D, Member 1/genetics , Adrenoleukodystrophy/genetics , Fibroblasts , Humans , Male , Mutation/genetics
7.
Asian Pac Isl Nurs J ; 5(2): 63-72, 2020.
Article in English | MEDLINE | ID: mdl-33043135

ABSTRACT

This study evaluated the effectiveness of a life-enhancement program designed to focus on dining conditions in welfare facilities for seniors living in Japan. Effectiveness was specifically evaluated based on whether improvements were achieved in (1) nutritional status, (2) oral health, (3) frequency of fever, and (4) vitality of appetite across three sites. As part of a comprehensive-care initiative that began with dining support, the program consisted of two main components: (1) a 3-month intensive program comprised of (a) collective experiential learning for residents and staff (including nutritionists, nurses, and physiotherapists) and (b) a tailor-made individual program for residents followed by (2) a 3-month continuation program. Participants included 168 individuals (31 males and 137 females) from a total of three facilities (average age was 85.9 [60-104] years). Results showed that the intensive program significantly improved nutritional status (e.g., BMI, caloric intake, and water intake; P < 0.000-0.005) and tongue movement (P < 0.000) while significantly reducing dental-plaque and tongue-coating indices (P < 0.000). Significant improvements were also achieved for degree of appetite and vitality indices (P < 0.000-0.001). However, incidences of fever were not reduced. These findings indicate that the program effectively improved nutritional status, oral health, vitality, and appetite. However, these effects did not sufficiently remain once the program was finished, thus suggesting the need for a continuous intervention.

8.
Front Cell Neurosci ; 14: 224, 2020.
Article in English | MEDLINE | ID: mdl-32922264

ABSTRACT

Chromosomal abnormality causes congenital and acquired intractable diseases. In general, there are no fundamental treatments for these diseases. To establish platforms to develop therapeutics for these diseases, patient-derived induced pluripotent stem cells (iPSCs) are highly beneficial. To study abnormal chromosomal diseases, it is often hard to apply animal disease models because the chromosomal structures are variable among species. It is also difficult to apply simple genome editing technology in cells or individuals for abnormal chromosomes. Thus, these patient-derived iPSCs have advantages for developing disease models with multiple cell and tissue types, which are typically seen in the symptoms of abnormal chromosomal diseases. Here we review the studies of patient-derived iPSCs carrying abnormal chromosomes, focusing on pluripotent state and neural lineages. We also discuss the technological advances in chromosomal manipulations toward establishing experimental models and future therapeutics. Patient-derived iPSCs carrying chromosomal abnormality are valuable as cellular bioresources since they can indefinitely proliferate and provide various cell types. Also, these findings and technologies are important for future studies on elucidating pathogenesis, drug development, regenerative medicine, and gene therapy for abnormal chromosomal diseases.

9.
Stem Cell Res ; 45: 101815, 2020 05.
Article in English | MEDLINE | ID: mdl-32361464

ABSTRACT

Juvenile nephronophthisis is an inherited renal ciliopathy, causing cystic kidney disease, renal fibrosis, and end-stage renal failure. Human induced pluripotent stem cell (hiPSC) lines, derived from two Juvenile nephronophthisis patients, were generated from peripheral blood mononuclear cells by episomal plasmid vectors. Generated hiPSC lines showed self-renewal and pluripotency and carried a large deletion in NPHP1 (Nephrocystin 1) gene. Since the molecular pathogenesis caused by NPHP1 dysfunction remains unclear, these cell resources provide useful tools to establish disease models and to develop new therapies for juvenile nephronophthisis.


Subject(s)
Induced Pluripotent Stem Cells , Adaptor Proteins, Signal Transducing , Cytoskeletal Proteins , Fibrosis , Humans , Kidney Diseases, Cystic/congenital , Leukocytes, Mononuclear , Membrane Proteins/genetics
10.
Sci Rep ; 7: 42186, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28176852

ABSTRACT

The tumour microenvironment is critical for various characteristics of tumour malignancies. Platelets, as part of the tumour microenvironment, are associated with metastasis formation via increasing the rate of tumour embolus formation in microvasculature. However, the mechanisms underlying the ability of tumour cells to acquire invasiveness and extravasate into target organs at the site of embolization remain unclear. In this study, we reported that platelet aggregation-inducing factor podoplanin expressed on tumour cell surfaces were found to not only promote the formation of tumour-platelet aggregates via interaction with platelets, but also induced the epithelial-mesenchymal transition (EMT) of tumour cells by enhancing transforming growth factor-ß (TGF-ß) release from platelets. In vitro and in vivo analyses revealed that podoplanin-mediated EMT resulted in increased invasiveness and extravasation of tumour cells. Treatment of mice with a TGF-ß-neutralizing antibody statistically suppressed podoplanin-mediated distant metastasis in vivo, suggesting that podoplanin promoted haematogenous metastasis in part by releasing TGF-ß from platelets that was essential for EMT of tumour cells. Therefore, our findings suggested that blocking the TGF-ß signalling pathway might be a promising strategy for suppressing podoplanin-mediated haematogenous metastasis in vivo.


Subject(s)
Carcinoma, Small Cell/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Membrane Glycoproteins/genetics , Transforming Growth Factor beta/genetics , A549 Cells , Animals , Antibodies, Neutralizing/pharmacology , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Platelets/pathology , Carcinoma, Small Cell/drug therapy , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/pathology , Cell Movement/drug effects , Diffusion Chambers, Culture , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred ICR , Neoplasm Invasiveness , Platelet Aggregation/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Tumor Microenvironment
11.
Oncotarget ; 7(4): 3934-46, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26684030

ABSTRACT

Podoplanin/Aggrus is a sialoglycoprotein expressed in various cancers. We previously identified podoplanin as a key factor in tumor-induced platelet aggregation. Podoplanin-mediated platelet aggregation enhances tumor growth and metastasis by secreting growth factors and by forming tumor emboli in the microvasculature. Thus, precise analysis of the mechanisms of podoplanin-mediated platelet aggregation is critical for developing anti-tumor therapies. Here we report the discovery of a novel platelet aggregation-inducing domain, PLAG4 (81-EDLPT-85). PLAG4 has high homology to the previously reported PLAG3 and contributes to the binding of its platelet receptor CLEC-2. Mutant analyses indicated that PLAG4 exhibits a predominant platelet-aggregating function relative to PLAG3 and that conserved Glu81/Asp82/Thr85 residues in PLAG4 are indispensable for CLEC-2 binding. By establishing anti-PLAG4-neutralizing monoclonal antibodies, we confirmed its role in CLEC-2 binding, platelet aggregation, and tumor emboli formation. Our results suggest the requirement of simultaneous inhibition of PLAG3/4 for complete suppression of podoplanin-mediated tumor growth and metastasis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Blood Platelets/pathology , Lectins, C-Type/metabolism , Lung Neoplasms/prevention & control , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Blood Platelets/drug effects , Blotting, Western , Female , Humans , Lectins, C-Type/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, Nude , Molecular Sequence Data , Platelet Aggregation Inhibitors/immunology , Protein Structure, Tertiary , Sequence Homology, Amino Acid
12.
Cancer Med ; 3(6): 1595-604, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25132683

ABSTRACT

Almost all highly metastatic tumor cells possess high platelet aggregating abilities, thereby form large tumor cell-platelet aggregates in the microvasculature. Embolization of tumor cells in the microvasculature is considered to be the first step in metastasis to distant organs. We previously identified the platelet aggregation-inducing factor expressed on the surfaces of highly metastatic tumor cells and named as Aggrus. Aggrus was observed to be identical to the marker protein podoplanin (alternative names, T1α, OTS-8, and others). Aggrus is frequently overexpressed in several types of tumors and enhances platelet aggregation by interacting with the platelet receptor C-type lectin-like receptor 2 (CLEC-2). Here, we generated a novel single-chain antibody variable region fragment (scFv) by linking the variable regions of heavy and light chains of the neutralizing anti-human Aggrus monoclonal antibody MS-1 with a flexible peptide linker. Unfortunately, the generated KM10 scFv failed to suppress Aggrus-induced platelet aggregation in vitro. Therefore, we performed phage display screening and finally obtained a high-affinity scFv, K-11. K-11 scFv was able to suppress Aggrus-induced platelet aggregation in vitro. Moreover, K-11 scFv prevented the formation of pulmonary metastasis in vivo. These results suggest that K-11 scFv may be useful as metastasis inhibitory scFv and is expected to aid in the development of preclinical and clinical examinations of Aggrus-targeted cancer therapies.


Subject(s)
Lung Neoplasms/blood , Lung Neoplasms/therapy , Membrane Glycoproteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Platelet Aggregation/drug effects , Single-Chain Antibodies/pharmacology , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Monoclonal, Murine-Derived/pharmacology , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Female , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Membrane Glycoproteins/blood , Membrane Glycoproteins/immunology , Membrane Proteins/blood , Membrane Proteins/immunology , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Rabbits , Single-Chain Antibodies/immunology
13.
Cancer Sci ; 105(8): 983-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24974736

ABSTRACT

The interactions of tumor cells with platelets contribute to the progression of tumor malignancy, and the expression levels of platelet aggregation-inducing factors positively correlate with the metastatic potential of osteosarcoma cells. However, it is unclear how tumor-platelet interaction contributes to the proliferation of osteosarcomas. We report here that osteosarcoma-platelet interactions induce the release of platelet-derived growth factor (PDGF) from platelets, which promotes the proliferation of osteosarcomas. Co-culture of platelets with MG63 or HOS osteosarcoma cells, which could induce platelet aggregation, enhanced the proliferation of each cell line in vitro. Analysis of phospho-antibody arrays revealed that co-culture of MG63 cells with platelets induced the phosphorylation of platelet derived growth factor receptor (PDGFR) and Akt. The addition of supernatants of osteosarcoma-platelet reactants also increased the growth of MG63 and HOS cells as well as the level of phosphorylated-PDGFR and -Akt. Sunitinib or LY294002, but not erlotinib, significantly inhibited the platelet-induced proliferation of osteosarcoma cells, indicating that PDGF released from platelets plays an important role in the proliferation of osteosarcomas by activating the PDGFR and then Akt. Our results suggest that inhibitors that specifically target osteosarcoma-platelet interactions may eradicate osteosarcomas.


Subject(s)
Blood Platelets/physiology , Bone Neoplasms/metabolism , Osteosarcoma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Coculture Techniques , Humans , Immunoblotting , Mice , Platelet Aggregation/physiology , Signal Transduction/physiology
14.
Int J Cancer ; 134(11): 2605-14, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24222607

ABSTRACT

Platelet aggregation-inducing factor Aggrus, also known as podoplanin, is associated with tumor malignancy by promoting hematogenous metastasis. Aggrus overexpression has been reported in some tumor tissues including lung, esophagus, head and neck and brain. We here found the frequent upregulation of aggrus mRNA in urinary bladder cancers using cancer tissue panels from various organs. Immunohistochemical analysis confirmed Aggrus protein expression in urinary bladder cancers and suggested a positive correlation between Aggrus expression and metastatic tendency in bladder cancers. Endogenous expression of Aggrus protein on the cell surface was found in the mouse bladder cancer MBT-2 cell line and human bladder cancer SCaBER cell lines. Knockdown of Aggrus expression in MBT-2 cells decreased their ability to induce platelet aggregation and form pulmonary metastasis in syngeneic mouse models. Knockdown of Aggrus expression in the human bladder cancer SCaBER cells also attenuated their ability to induce platelet aggregation and form pulmonary metastasis in mice. Moreover, pulmonary metastasis of SCaBER cells was prevented by prior administration of our generated anti-Aggrus neutralizing monoclonal antibodies by attenuating their retention in lung. These results indicate that Aggrus plays an important role in bladder cancer metastasis. Thus, anti-Aggrus neutralizing antibodies would be useful for the prevention of hematogenous metastasis of Aggrus-positive bladder cancer.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell/metabolism , Carcinoma, Transitional Cell/metabolism , Lung Neoplasms/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Urinary Bladder Neoplasms/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , CHO Cells , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/pathology , Cell Proliferation , Cricetinae , Cricetulus , Flow Cytometry , Humans , Immunoenzyme Techniques , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Male , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred C3H , Mice, Inbred ICR , Platelet Aggregation , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
15.
PLoS One ; 8(8): e73609, 2013.
Article in English | MEDLINE | ID: mdl-23991201

ABSTRACT

The platelet aggregation-inducing factor Aggrus, also known as podoplanin, is frequently upregulated in several types of tumors and enhances hematogenous metastasis by interacting with and activating the platelet receptor CLEC-2. Thus, Aggrus-CLEC-2 binding could be a therapeutic molecular mechanism for cancer therapy. We generated a new anti-human Aggrus monoclonal antibody, MS-1, that suppressed Aggrus-CLEC-2 binding, Aggrus-induced platelet aggregation, and Aggrus-mediated tumor metastasis. Interestingly, the MS-1 monoclonal antibody attenuated the growth of Aggrus-positive tumors in vivo. Moreover, the humanized chimeric MS-1 antibody, ChMS-1, also exhibited strong antitumor activity against Aggrus-positive lung squamous cell carcinoma xenografted into NOD-SCID mice compromising antibody-dependent cellular cytotoxic and complement-dependent cytotoxic activities. Because Aggrus knockdown suppressed platelet-induced proliferation in vitro and tumor growth of the lung squamous cell carcinoma in vivo, Aggrus may be involved in not only tumor metastasis but also tumor growth by promoting platelet-tumor interaction, platelet activation, and secretion of platelet-derived factors in vivo. Our results indicate that molecular target drugs inhibiting specific platelet-tumor interactions can be developed as antitumor drugs that suppress both metastasis and proliferation of tumors such as lung squamous cell carcinoma.


Subject(s)
Blood Platelets/physiology , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Neoplasm Metastasis , Neoplasms/pathology , Animals , CHO Cells , Cricetinae , Cricetulus , Female , Mice , Mice, Inbred BALB C , Protein Binding
16.
Cancer Sci ; 102(11): 2051-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21824222

ABSTRACT

The platelet aggregation-inducing factor, Aggrus (also known as podoplanin), is reported to contribute to cancer metastasis by mediating cancer cell-platelet interaction. Aggrus has been shown to be upregulated in many different types of cancers. Thus, not only the functional inhibition of Aggrus, but also its application as a cancer-specific antigen has therapeutic potential. Among a series of anti-Aggrus mAb established previously, no mouse anti-human Aggrus mAb exists that possesses the ability to neutralize platelet aggregation. For precise preclinical examinations of mouse and monkey models, the establishment of Aggrus-neutralizing mouse mAb and their chimeric Abs is needed. In this study, we established two mouse anti-human Aggrus mAb, P2-0 and HAG-3. A precise analysis of their epitopes revealed that P2-0 recognized the conformation near the bioactive O-glycosylation site at the Thr(52) residue. In contrast, HAG-3 recognized the amino-terminus side at a short distance from the conformation recognized by P2-0. We observed that only P2-0 attenuated Aggrus-induced platelet aggregation and Aggrus binding to its platelet receptor, that is, the C-type lectin-like receptor-2. Consistent with these data, only P2-0 prevented the experimental metastasis of human Aggrus-overexpressing CHO cells. Subsequently, we cloned the complementary determining region of P2-0 and produced the murine/human chimeric P2-0 antibody. This chimeric antibody maintained its inhibitory activity of Aggrus-induced platelet aggregation and experimental metastasis. Thus, P2-0 and its chimeric antibody are expected to aid the development of preclinical and clinical examinations of Aggrus-targeted cancer therapy.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Neoplasm/immunology , Antibodies, Neutralizing/immunology , Epitopes/immunology , Membrane Glycoproteins/immunology , Neoplasm Metastasis/prevention & control , Neoplasm Proteins/immunology , Neoplastic Cells, Circulating/immunology , Platelet Aggregation/immunology , Animals , CHO Cells/pathology , Cricetinae , Cricetulus , Female , Humans , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Conformation , Recombinant Fusion Proteins/immunology , Species Specificity , Xenograft Model Antitumor Assays
17.
J Biol Chem ; 286(4): 2681-8, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21062737

ABSTRACT

The incidence and death rate of prostate cancer is increasing rapidly. In addition, the low sensitivity of prostate cancer to chemotherapy makes it difficult to treat this condition. The serine/threonine kinase Pim-1 plays an important role in cell cycle progression and apoptosis inhibition, resulting in prostate tumorigenesis. Therefore, Pim-1 inhibition has been expected to be an attractive target for developing new anti-cancer drugs. However, no small compounds targeting Pim-1 have progressed to clinical use because of their lack of specificity. Here, we have reported a new cell-permeable Pim-1 inhibitory p27(Kip1) peptide that could interfere with the binding of Pim-1 to its substrates and act as an anti-cancer drug. The peptide could bind to Pim-1 and inhibit phosphorylation of endogenous p27(Kip1) and Bad by Pim-1. Treatment of prostate cancer with the peptide induces G(1) arrest and subsequently apoptosis in vitro. However, the peptide showed almost no growth inhibitory or apoptosis-inducing effects in normal cells. The peptide could inhibit tumor growth in in vivo prostate cancer xenograft models. Moreover, the peptide treatment could overcome resistance to taxol, one of the first line chemotherapeutic agents for prostate cancer, and a combination of the peptide with taxol synergistically inhibited prostate cancer growth in vivo. These results indicate that a Pim-1 inhibitory p27(Kip1) peptide could be developed as an anti-cancer drug against prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Intracellular Signaling Peptides and Proteins/pharmacology , Peptides/pharmacology , Prostatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27 , Drug Screening Assays, Antitumor , G1 Phase/drug effects , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Peptides/metabolism , Phosphorylation , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins c-pim-1/metabolism , bcl-Associated Death Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...