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











Publication year range
1.
Int J Cancer ; 152(7): 1425-1437, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36412556

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is resistant to current treatments but lectin-based therapy targeting cell surface glycans could be a promising new horizon. Here, we report a novel lectin-based phototherapy (Lec-PT) that combines the PDAC targeting ability of rBC2LCN lectin to a photoabsorber, IRDye700DX (rBC2-IR700), resulting in a novel and highly specific near-infrared, light-activated, anti-PDAC therapy. Lec-PT cytotoxicity was first verified in vitro with a human PDAC cell line, Capan-1, indicating that rBC2-IR700 is only cytotoxic upon cellular binding and exposure to near-infrared light. The therapeutic efficacy of Lec-PT was subsequently verified in vivo using cell lines and patient-derived, subcutaneous xenografting into nude mice. Significant accumulation of rBC2-IR700 occurs as early as 2 hours postintravenous administration while cytotoxicity is only achieved upon exposure to near-infrared light. Repeated treatments further slowed tumor growth. Lec-PT was also assessed for off-target toxicity in the orthotopic xenograft model. Shielding of intraperitoneal organs from near-infrared light minimized off-target toxicity. Using readily available components, Lec-PT specifically targeted pancreatic cancer with high reproducibility and on-target, inducible toxicity. Rapid clinical development of this method is promising as a new modality for treatment of pancreatic cancer.


Subject(s)
Lectins , Pancreatic Neoplasms , Animals , Mice , Humans , Mice, Nude , Reproducibility of Results , Immunotherapy/methods , Cell Line, Tumor , Phototherapy/methods , Pancreatic Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Photosensitizing Agents/therapeutic use , Pancreatic Neoplasms
2.
Cancer Sci ; 112(9): 3722-3731, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34115906

ABSTRACT

The rBC2LCN lectin, known as a stem cell marker probe that binds to an H type 3 fucosylated trisaccharide motif, was recently revealed to also bind to pancreatic ductal adenocarcinoma (PDAC) cells. A lectin-drug conjugate was generated by fusing rBC2LCN with a cytocidal toxin, and it showed a strong anticancer effect in in vitro and in vivo PDAC models. However, it is unclear which molecules are carrier proteins of rBC2LCN on PDAC cells. In this study, we identified a rBC2LCN-positive glycoprotein expressed in PDAC. Tumor lysates of PDAC patient-derived xenografts (PDXs) were coprecipitated with rBC2LCN lectin and analyzed by liquid chromatography-mass spectrometry. A total of 343 proteins were initially identified. We used a web-based database to select five glycoproteins and independently evaluated their expression in PDAC by immunohistochemistry (IHC). Among them, we focused on carcinoembryonic antigen 5 (CEA) as the most cancer-specific carrier protein in PDAC, as it showed the most prominent difference in expression rate between PDAC cells (74%) and normal pancreatic duct cells (0%, P > .0001). rBC2LCN lectin and CEA colocalization in PDAC samples was confirmed by double-staining analysis. Furthermore, rBC2LCN-precipitated fractions were blotted with an anti-CEA polyclonal antibody (pAb), and CEA pAb-precipitated fractions were blotted with rBC2LCN lectin. The results demonstrate that CEA is in fact a ligand of rBC2LCN lectin.


Subject(s)
Carcinoembryonic Antigen/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carrier Proteins/metabolism , Lectins/metabolism , Pancreatic Neoplasms/metabolism , Animals , Antibodies/immunology , Carcinoembryonic Antigen/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Female , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Heterografts , Humans , Immunohistochemistry/methods , Immunoprecipitation/methods , Ligands , Mice , Mice, Inbred ICR , Mice, SCID , Neoplasm Transplantation , Pancreatic Neoplasms/pathology
3.
Biochem Biophys Res Commun ; 534: 310-316, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33288196

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an intractable malignancy for which novel therapeutic targets are in high demand. To uncover glycans expressed within PDAC, we previously performed glycome profiling of PDAC cell lines using lectin microarray and found that the lectin rBC2LCN with specificity to a Fucα1-2Galß1-3 motif exhibited strong binding to a PDAC cell line (Capan-1) and to all tumor tissues derived from 69 pancreatic cancer patients. Nevertheless, no information was available as to whether glycans containing the Fucα1-2Galß1-3 motif are expressed within PDAC. Here we used HPLC combined with MALDI-TOFMS to perform a structural and quantitative glycome analysis targeting both N- and O-glycans derived from two types of patient-derived PDAC xenograft mouse models, PC3 (well-differentiated) and PC42 (poorly-differentiated). A higher percentage of highly branched and sialylated complex-type N-glycans was detected in PC42 relative to PC3. The percentage of core 1 O-glycans was higher in PC42 relative to PC3, whereas that of core 3 O-glycans was higher in PC3. Cancer-related glycan epitopes such as Lewis A and Lewis Y were detected in core 3 O-glycans of both PC3 and PC42. H-type3 containing the Fucα1-2Galß1-3 motif was detected in Core 2 O-glycans in both models, explaining the molecular mechanism of the binding of rBC2LCN to PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/chemistry , Pancreatic Neoplasms/chemistry , Polysaccharides/chemistry , Animals , Carbohydrate Conformation , Carbohydrate Sequence , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Differentiation , Cell Line, Tumor , Female , Glycomics , Heterografts , Humans , Mice , Mice, SCID , Molecular Structure , Neoplasm Transplantation , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Polysaccharides/metabolism
4.
Cancer Sci ; 111(12): 4548-4557, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33058342

ABSTRACT

Drug resistance represents an obstacle in colorectal cancer (CRC) treatment because of its association with poor prognosis. rBC2LCN is a lectin isolated from Burkholderia that binds cell surface glycans that have fucose moieties. Because fucosylation is enhanced in many types of cancers, this lectin could be an efficient drug carrier if CRC cells specifically present such glycans. Therefore, we examined the therapeutic efficacy and toxicity of lectin drug conjugate therapy in CRC mouse xenograft models. The affinity of rBC2LCN for human CRC cell lines HT-29, LoVo, LS174T, and DLD-1 was assessed in vitro. The cytocidal efficacy of a lectin drug conjugate, rBC2LCN-38 kDa domain of pseudomonas exotoxin A (PE38) was evaluated by MTT assay. The therapeutic effects and toxicity for each CRC cell line-derived mouse xenograft model were compared between the intervention and control groups. LS174T and DLD-1 cell lines showed a strong affinity for rBC2LCN. In the xenograft model, the tumor volume in the rBC2LCN-PE38 group was significantly reduced compared with that using control treatment alone. However, the HT-29 cell line showed weak affinity and poor therapeutic efficacy. No significant toxicities or adverse responses were observed. In conclusion, we demonstrated that rBC2LCN lectin binds CRC cells and that rBC2LCN-PE38 significantly suppresses tumor growth in vivo. In addition, the efficacy of the drug conjugate correlated with its binding affinity for each CRC cell line. These results suggest that lectin drug conjugate therapy has potential as a novel targeted therapy for CRC cell surface glycans.


Subject(s)
ADP Ribose Transferases/therapeutic use , Adenocarcinoma/drug therapy , Bacterial Toxins/therapeutic use , Colorectal Neoplasms/drug therapy , Exotoxins/therapeutic use , Immunoconjugates/therapeutic use , Lectins/therapeutic use , Virulence Factors/therapeutic use , ADP Ribose Transferases/adverse effects , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Bacterial Toxins/adverse effects , Burkholderia cenocepacia/chemistry , Cell Line, Tumor , Cell Survival , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Carriers , Exotoxins/adverse effects , Fucose/metabolism , Fucosyltransferases/metabolism , HT29 Cells , Heterografts , Humans , Immunoconjugates/adverse effects , In Vitro Techniques , Lectins/isolation & purification , Lectins/metabolism , Mice , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/therapeutic use , Tumor Burden , Virulence Factors/adverse effects , Pseudomonas aeruginosa Exotoxin A
5.
Biochem Biophys Res Commun ; 532(4): 647-654, 2020 11 19.
Article in English | MEDLINE | ID: mdl-32912628

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) are useful starting materials for the generation of cell therapy products, due to their pluripotency and ability to self-renew. Quality control of hiPSCs is extremely important in creating a stable supply of hPSC-derived products. Previously we identified an hiPSC-specific lectin probe, rBC2LCN, which binds specifically to α1,2-fucosylated glycan and recognizes podocalyxin (PODXL) as a glycoprotein ligand. In this study, we produced monoclonal antibodies (mAbs) specific for α1,2-fucosylated PODXL expressed on hiPSCs. PODXL was recombinantly expressed in fucosyltransferase 1 (FUT1)-transfected HEK293, followed by immunization into mice. Monoclonal antibodies, which bind to PODXL/FUT1-transfected cells, but not to cells transfected with only one of PODXL or FUT1, were screened by flow cytometry. The two mAbs generated (179-6B8C9 and 179-7E12E10), termed α1,2-fucosylated PODXL-specific mAbs (FpMabs), showed binding specificity to PODXL/FUT1-transfected cells. The FpMabs bound to hiPSCs but never to human adipose-derived mesenchymal stem cells, human dermal fibroblasts, or hiPSC-derived mesoderm. Altogether, FpMabs are highly specific probes for hiPSCs, which might be a powerful tool for the characterization of hiPSCs used in regenerative medicine.


Subject(s)
Antibodies, Monoclonal , Induced Pluripotent Stem Cells/immunology , Sialoglycoproteins/immunology , Animals , Cell Line , Cells, Cultured , Flow Cytometry , Fluorescence , Fucosyltransferases/genetics , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Polysaccharides/analysis , Polysaccharides/immunology , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Transfection
6.
Regen Ther ; 14: 306-314, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32462059

ABSTRACT

INTRODUCTION: Tumourigenesis attributed to residual undifferentiated cells in a graft is considered to be a significant issue in cell therapy using human pluripotent stem cells. To ensure the safety of regenerative medicine derived from pluripotent stem cells, residual undifferentiated cells must be eliminated in the manufacturing process. We previously described the lectin probe rBC2LCN, which binds harmlessly and specifically to the cell surface of human pluripotent stem cells. We report here a technique using rBC2LCN to remove pluripotent cells from a heterogenous population to reduce the chance of teratoma formation. METHODS: We demonstrate a method for separating residual tumourigenic cells using rBC2LCN-bound magnetic beads. This technology is a novel use of their previous discovery that rBC2LCN is a lectin that selectively binds to pluripotent cells. We optimize and validate a method to remove hPSCs from a mixture with human fibroblasts using rBC2LCN-conjugated magnetic beads. RESULTS: Cells with the potential to form teratoma could be effectively eliminated from a heterogeneous cell population with biotin-labelled rBC2LCN and streptavidin-bound magnetic beads. The efficiency was measured by FACS, ddPCR, and animal transplantation, suggesting that magnetic cell separation using rBC2LCN is quite efficient for eliminating hPSCs from mixed cell populations. CONCLUSIONS: The removal of residual tumourigenic cells based on rBC2LCN could be a practical option for laboratory use and industrialisation of regenerative medicine using human pluripotent stem cells.

7.
Methods Mol Biol ; 2132: 453-461, 2020.
Article in English | MEDLINE | ID: mdl-32306352

ABSTRACT

rBC2LCN is a recombinant N-terminal domain of BC2L-C lectin that is derived from the gram-negative bacteria Burkholderia cenocepacia and specifically binds to Fucα1-2Galß1-3GlcNAc/GalNAc. Glycome analysis using a high-density lectin microarray revealed that rBC2LCN specifically binds to human pluripotent stem cells (hPSCs) but not to non-hPSCs. The lectin can be added to the cell culture medium for the live staining of hPSCs without causing visible cytotoxicity. Moreover, it can be used in flow cytometric analysis and for the staining of fixed hPSCs. rBC2LCN is a single-chain molecule with a low molecular weight (trimer of 16 kDa), which can be produced in large quantities in Escherichia coli (0.1 g/L). Therefore, rBC2LCN may be a cost-effective probe for use in hPSCs, unlike other hPSC surface marker antibodies that require a mammalian cell expression system for production. In this study, we describe the protocols for the expression and purification of rBC2LCN from E. coli and live staining of hPSCs using this probe.


Subject(s)
Burkholderia cenocepacia/metabolism , Escherichia coli/growth & development , Lectins/chemistry , Lectins/genetics , Pluripotent Stem Cells/cytology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Culture Techniques , Cells, Cultured , Escherichia coli/genetics , Flow Cytometry , Glycomics/methods , Humans , Lectins/metabolism , Molecular Probes/metabolism , Pluripotent Stem Cells/metabolism , Protein Domains , Protein Engineering , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
8.
FEBS Open Bio ; 10(6): 1056-1064, 2020 06.
Article in English | MEDLINE | ID: mdl-32237061

ABSTRACT

The recombinant N-terminal domain of BC2L-C lectin (rBC2LCN) is useful for detecting not only human pluripotent stem cells but also some cancers. However, the cancer types and stages that can be detected by rBC2LCN remain unclear. In this study, we identified the human breast carcinoma subtypes and stages that can be detected by rBC2LCN. Compared with rBC2LCN-negative breast carcinoma cell lines, the rBC2LCN-positive cells expressed higher levels of human epidermal growth factor receptor 2 (HER2) and epithelial marker genes. Importantly, rBC2LCN histochemical staining of human breast carcinoma tissues demonstrated the utility of rBC2LCN in detecting breast carcinoma types that express HER2 and have not spread much in the early phase of growth. We conclude that rBC2LCN may have potential as a detection probe and a drug delivery vehicle to identify and treat early-stage HER2-positive breast carcinoma.


Subject(s)
Bacterial Proteins/chemistry , Breast Neoplasms/diagnosis , Lectins/chemistry , Molecular Probes/chemistry , Antineoplastic Agents/administration & dosage , Bacterial Proteins/genetics , Breast/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Burkholderia cenocepacia , Drug Carriers/chemistry , Feasibility Studies , Female , Humans , Lectins/genetics , MCF-7 Cells , Molecular Probes/genetics , Neoplasm Staging , Receptor, ErbB-2/analysis , Receptor, ErbB-2/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Tissue Array Analysis/methods
9.
Eur Surg Res ; 61(4-5): 113-122, 2020.
Article in English | MEDLINE | ID: mdl-33503609

ABSTRACT

INTRODUCTION: Since the outermost layer of cancer cells is covered with various glycans, targeting these groups may serve as an effective strategy in cancer therapy. We previously reported that fucosylated glycans are specifically expressed on pancreatic cancer cells, and that a protein specifically binding to these glycans, namely rBC2LCN lectin, is a potential guiding drug carrier. In the present study, a novel type of glycan-targeting nanoparticle was developed by modifying the surface of doxorubicin-containing liposomes with rBC2LCN lectin. The efficiency and specificity of this formulation, termed Lec-Doxosome, were examined in vitro and in vivo in human pancreatic cancer models. METHODS: Lec-Doxosome was prepared by a post-insertion method based on the insertion of rBC2LCN lectin into the liposomal surface via a lipid linker. The in vitro cellular binding, uptake, and cytotoxicity of Lec-Doxosome were compared with the corresponding parameters in the unmodified liposomes by applying to human pancreatic cancer cell line (Capan-1) with affinity for rBC2LCN lectin. For the in vivo assay, Lec-Doxosome was intravenously injected once per week for a total of 3 weeks into mice bearing subcutaneous tumors. RESULTS: The in vitro application of Lec-Doxosome resulted in a 1.2- to 1.6-fold higher intracellular doxorubicin accumulation and a 1.5-fold stronger cytotoxicity compared with the respective rates of accumulation and cytotoxicity in the unmodified liposomes. In vivo, Lec-Doxosome reduced the mean tumor weight (368 mg) compared with that in mice treated with unmodified liposomes (456 mg), without causing any additional adverse events. CONCLUSION: It was demonstrated from the results obtained herein that rBC2LCN lectin is a potent modifier, as a means for boosting the efficiency of nanoparticles in the targeting of cancer cell surface glycans.


Subject(s)
Doxorubicin/analogs & derivatives , Drug Delivery Systems , Lectins/chemistry , Pancreatic Neoplasms/drug therapy , Polysaccharides/metabolism , Animals , Cell Line, Tumor , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Doxorubicin/metabolism , Female , Humans , Lectins/metabolism , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Polyethylene Glycols/metabolism
10.
Biochem Biophys Res Commun ; 515(1): 176-182, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31133376

ABSTRACT

The recombinant lectin rBC2LCN is a useful marker for discriminating the undifferentiated status of human induced or embryonic stem cells. Recently, rBC2LCN has also been used for detecting some cancers and niche cells. However, the generality of which types of cells are detected by rBC2LCN is unclear. In this study, we demonstrated the potential of rBC2LCN as a probe for detecting and isolating cancer stem-like cells. Interestingly, flow cytometric analysis of various human cell lines indicated that the human prostate cancer cell line PC-3 consisted of rBC2LCN-positive and -negative subpopulations. Compared with the rBC2LCN-negative subpopulation, the rBC2LCN-positive subpopulation possessed representative features of cancer stem cells and malignancy, such as slow proliferation, increased cell motility, anchorage-independent growth, and drug resistance. The comprehensive expression profiles revealed that the rBC2LCN-positive subpopulation expressed higher levels of cancer stem cell markers. These findings indicate that rBC2LCN is useful for detecting not only pluripotent stem cells but also the cancer stem-like subpopulation of PC-3 cells. Pluripotent and cancer cells with rBC2LCN positivity would be important for future stem cell research.


Subject(s)
Lectins/metabolism , Molecular Probes/metabolism , Neoplastic Stem Cells/metabolism , Prostatic Neoplasms/metabolism , Recombinant Proteins/metabolism , Biomarkers, Tumor/genetics , Cell Line , Cell Line, Tumor , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Human Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Lectins/genetics , MCF-7 Cells , Male , Molecular Probes/genetics , Neoplastic Stem Cells/pathology , PC-3 Cells , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
11.
Comp Biochem Physiol A Mol Integr Physiol ; 166(4): 590-602, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24036479

ABSTRACT

Satellite cells are multipotential stem cells responsible for muscle growth and regeneration. Satellite cell proliferation, differentiation, and responsiveness to fibroblast growth factor 2 (FGF2) is, in part, regulated by the heparan sulfate proteoglycans syndecan-4 and glypican-1. Syndecan-4 and glypican-1 expression declines with satellite cell age and may be associated with decreased satellite cell activity. The objective of the current study was to determine if overexpression of syndecan-4 and glypican-1 would increase proliferation, differentiation and FGF2 responsiveness in satellite cells isolated from pectoralis major muscle from 16-wk-old turkeys. Overexpression of syndecan-4 and glypican-1 did not have a significant effect on proliferation and differentiation in 1d, 7 wk, and 16 wk satellite cells, and did not affect FGF2 responsiveness during proliferation. Expression of syndecan-4 and glypican-1 increased differentiation at 48 h in 1d, 7 wk, and 16 wk cells treated with FGF2. Expression of myogenic regulatory factors MyoD, myogenin, and MRF4 was affected by the overexpression of syndecan-4 and glypican-1. However, changes in myogenic regulatory factor expression did not have a significant effect on proliferation or differentiation. These data demonstrate that syndecan-4 and glypican-1 are likely not directly associated with the age related decrease in satellite cell activity.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Glypicans/biosynthesis , Satellite Cells, Skeletal Muscle/physiology , Syndecan-4/biosynthesis , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cellular Senescence , Male , Satellite Cells, Skeletal Muscle/drug effects , Turkeys
SELECTION OF CITATIONS
SEARCH DETAIL