Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(6): e0269547, 2022.
Article in English | MEDLINE | ID: mdl-35679330

ABSTRACT

Human pluripotent stem cells (hPSCs) exist in at least two distinct states in mammals: naïve pluripotency that represents several molecular characteristics in pre-implantation epiblast and primed pluripotency that corresponds to cells poised for differentiation in post-implantation epiblast. To identify and characterize the surface molecules that are necessary for the maintenance of naïve hPSCs, we generated a panel of murine monoclonal antibodies (MAbs) specific to the naïve state of hPSCs. Flow cytometry showed that N1-A4, one of the MAbs, bound to naïve hPSCs but not to primed hPSCs. Cell surface biotinylation and immunoprecipitation analysis identified that N1-A4 recognized heat shock protein 60 (HSP60) expressed on the surface of naïve hPSCs. Quantitative polymerase chain reaction (qPCR) analysis showed that HSP60 expression was rapidly downregulated during the embryoid body (EB) differentiation of primed hPSCs. HSP60 knockdown led to a decrease in the expression of pluripotency genes in primed hPSCs. HSP60 depletion also led to a decrease in the expression of pluripotency genes and representative naïve-state-specific genes in naïve hPSCs. Taken together, the results suggest that HSP60 is downregulated during differentiation of hPSCs and is required for the maintenance of pluripotency genes in both primed and naïve hPSCs, suggesting that HSP60 is a regulator of hPSC pluripotency and differentiation.


Subject(s)
Chaperonin 60 , Pluripotent Stem Cells , Animals , Cell Differentiation , Chaperonin 60/genetics , Chaperonin 60/metabolism , Embryoid Bodies , Germ Layers , Humans , Mammals , Mice
2.
Oncol Rep ; 46(2)2021 Aug.
Article in English | MEDLINE | ID: mdl-34109989

ABSTRACT

Lung cancer is the most frequent cause of cancer­associated mortality worldwide. Upregulation of heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNPA2/B1) has been reported in non­small cell lung cancer (NSCLC) cells, but its contribution to NSCLC remains poorly understood. hnRNPA2/B1 is involved in carcinogenesis by interacting with a number of proteins; however, little is known about its interaction with p53. The results of the present study revealed that hnRNPA2/B1 expression levels were upregulated in NSCLC cells under tumorsphere culture conditions and cisplatin treatment compared with those in cells under the adherent condition and dimethyl sulfoxide treatment, respectively, suggesting that hnRNPA2/B1 expression is induced under stress conditions. hnRNPA2/B1 knockdown decreased the number and size of NSCLC cell colonies in a clonogenic survival assay and led to a decreased migratory potential of NSCLC cells, suggesting that hnRNPA2/B1 may promote the survival, proliferation and migration of NSCLC cells. hnRNPA2/B1 knockdown induced G0/G1 phase arrest in NSCLC cells through cyclin E degradation and phosphorylation of cyclin­dependent kinase 2. In addition, hnRNPA2/B1 knockdown inhibited extracellular signal­regulated kinase (ERK)1/2 phosphorylation, suggesting that hnRNPA2/B1 may promote the G1/S phase transition in NSCLC cells through ERK signaling. hnRNPA2/B1 knockdown resulted in increased expression levels of p21 and p27 in NSCLC cells, as well as p53 induction and phosphorylation. Additionally, hnRNPA2/B1 knockdown inhibited human double minute 2 protein (HDM2) stability and phosphorylation, whereas overexpression of hnRNPA2 induced the opposite effects. These results suggested that hnRNPA2/B1 may promote the survival, proliferation and migration of NSCLC cells through preventing the activation of p53, which is induced by ERK­mediated HDM2 activation. The results of the present study also indicated that the components of the hnRNPA2/B1/ERK/p53/HDM2 signaling pathway may be novel potential molecular targets for the treatment of patients with NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cisplatin/pharmacology , Dimethyl Sulfoxide/pharmacology , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Lung Neoplasms/metabolism , A549 Cells , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , MAP Kinase Signaling System/drug effects , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism
3.
Stem Cell Res Ther ; 10(1): 328, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31744543

ABSTRACT

Previous studies including ours have demonstrated a critical function of the transcription factor ETV2 (ets variant 2; also known as ER71) in determining the fate of cardiovascular lineage development. However, the underlying mechanisms of ETV2 function remain largely unknown. In this study, we demonstrated the novel function of the miR (micro RNA)-126-MAPK (mitogen-activated protein kinase) pathway in ETV2-mediated FLK1 (fetal liver kinase 1; also known as VEGFR2)+ cell generation from the mouse embryonic stem cells (mESCs). By performing a series of experiments including miRNA sequencing and ChIP (chromatin immunoprecipitation)-PCR, we found that miR-126 is directly induced by ETV2. Further, we identified that miR-126 can positively regulate the generation of FLK1+ cells by activating the MAPK pathway through targeting SPRED1 (sprouty-related EVH1 domain containing 1). Further, we showed evidence that JUN/FOS activate the enhancer region of FLK1 through AP1 (activator protein 1) binding sequences. Our findings provide insight into the novel molecular mechanisms of ETV2 function in regulating cardiovascular lineage development from mESCs.


Subject(s)
MAP Kinase Signaling System , MicroRNAs/metabolism , Mouse Embryonic Stem Cells/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Binding Sites , Calcium-Binding Proteins/genetics , EGF Family of Proteins/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Mice , Promoter Regions, Genetic/genetics
4.
Cancer Lett ; 454: 44-52, 2019 07 10.
Article in English | MEDLINE | ID: mdl-30980864

ABSTRACT

Hepatocellular carcinoma (HCC) is currently the third leading cause of cancer death worldwide. To study how mycoplasma infection affects HCC progression, we investigated the characteristics of mycoplasma-infected tumor tissues and circulating tumor cells (CTCs) in HCC patients. The mycoplasmal membrane protein p37 showed significant correlations with higher histologic stages and vascular invasion and predicted poor disease-free survival of HCC patients. p37-positive CTCs were detected in 42 out of 47 HCC patients (89%). p37-positive circulating cells were also detected in 4 out of 10 healthy donors (40%), and all were epithelial cell adhesion molecule (EpCAM)-positive. In HCC patients, most of p37-negative CTCs (95%) showed intermediate phenotype with neither EpCAM nor vimentin expression, but p37-positive CTCs were EpCAM-positive (44%), vimentin-positive (32%), and both negative (24%), suggesting that EpCAM-positive CTCs are enriched with mycoplasma infection. Mycoplasma infection promoted migratory capacity of HCC cells with increased expression of EpCAM. Immunoprecipitation analysis revealed that p37 associates with EpCAM. The results suggest that mycoplasma infection promotes tumor progression in HCC patients via interaction of the mycoplasmal p37 and EpCAM.


Subject(s)
Antigens, Bacterial/metabolism , Carcinoma, Hepatocellular/microbiology , Epithelial Cell Adhesion Molecule/metabolism , Liver Neoplasms/microbiology , Mycoplasma Infections/metabolism , Mycoplasma hyorhinis/metabolism , A549 Cells , Antigens, Bacterial/biosynthesis , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Disease Progression , Hep G2 Cells , Humans , Immunohistochemistry , Liver Neoplasms/blood , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mycoplasma Infections/blood , Mycoplasma Infections/microbiology , Mycoplasma Infections/pathology , Neoplastic Cells, Circulating
5.
Sci Rep ; 9(1): 653, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679694

ABSTRACT

Progesterone receptor membrane component1 (PGRMC1) is a heme-binding protein involved in cancers and Alzheimer's disease. PGRMC1 consists of a short N-terminal extracellular or luminal domain, a single membrane-spanning domain, and a long cytoplasmic domain. Previously, we generated two monoclonal antibodies (MAbs) 108-B6 and 4A68 that recognize cell surface-expressed PGRMC1 (csPGRMC1) on human pluripotent stem cells and some cancer cells. In this study, flow cytometric analysis found that an anti-PGRMC1 antibody recognizing the N-terminus of PGRMC1 could not bind to csPGRMC1 on cancer cells, and 108-B6 and 4A68 binding to csPGRMC1 was inhibited by trypsin treatment, suggesting that the epitopes of 108-B6 and 4A68 are trypsin-sensitive. To examine the epitope specificity of 108-B6 and 4A68, glutathione-S-transferase (GST)-fused PGRMC1 mutants were screened to identify the epitopes targeted by the antibodies. The result showed that 108-B6 and 4A68 recognized C-terminal residues 183-195 and 171-182, respectively, of PGRMC1, where trypsin-sensitive sites are located. A polyclonal anti-PGRMC1 antibody raised against the C-terminus of PGRMC1 could also recognized csPGRMC1 in a trypsin-sensitive manner, suggesting that the C-terminus of csPGRMC1 is exposed on the cell surface. This finding reveals that csPGRMC1 has a non-conventional plasma membrane topology, which is different from that of intracellular PGRMC1.


Subject(s)
Antibodies, Monoclonal/immunology , Cell Membrane/metabolism , Epitope Mapping , Membrane Proteins/immunology , Membrane Proteins/metabolism , Receptors, Progesterone/immunology , Receptors, Progesterone/metabolism , Cell Line , Humans , Intracellular Space/metabolism , Membrane Proteins/chemistry , Receptors, Progesterone/chemistry
6.
Sci Rep ; 8(1): 3048, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29445107

ABSTRACT

Progesterone receptor membrane component 1 (PGRMC1) is a multifunctional heme-binding protein involved in various diseases, including cancers and Alzheimer's disease. Previously, we generated two monoclonal antibodies (MAbs) 108-B6 and 4A68 against surface molecules on human pluripotent stem cells (hPSCs). Here we show that PGRMC1 is the target antigen of both MAbs, and is predominantly expressed on hPSCs and some cancer cells. PGRMC1 is rapidly downregulated during early differentiation of hPSCs. Although PGRMC1 knockdown leads to a spread-out morphology and impaired self-renewal in hPSCs, PGRMC1 knockdown hPSCs do not show apoptosis and autophagy. Instead, PGRMC1 knockdown leads to differentiation of hPSCs into multiple lineage cells without affecting the expression of pluripotency markers. PGRMC1 knockdown increases cyclin D1 expression and decreases Plk1 expression in hPSCs. PGRMC1 knockdown also induces p53 expression and stability, suggesting that PGRMC1 maintains hPSC self-renewal through suppression of p53-dependent pathway. Analysis of signaling molecules further reveals that PGRMC1 knockdown promotes inhibitory phosphorylation of GSK-3ß and increased expression of Wnt3a and ß-catenin, which leads to activation of Wnt/ß-catenin signaling. The results suggest that PGRMC1 suppresses the p53 and Wnt/ß-catenin pathways to promote self-renewal and inhibit early differentiation in hPSCs.


Subject(s)
Cell Self Renewal/physiology , Membrane Proteins/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Receptors, Progesterone/metabolism , Wnt Signaling Pathway/physiology , Apoptosis/physiology , Cell Differentiation/physiology , Cell Lineage , Cell Proliferation/physiology , Down-Regulation , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , Human Embryonic Stem Cells , Humans , Phosphorylation , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Wnt3A Protein/metabolism , beta Catenin/metabolism
7.
PLoS One ; 12(11): e0188075, 2017.
Article in English | MEDLINE | ID: mdl-29145450

ABSTRACT

B-cell receptor-associated protein 31 (BAP31) is an endoplasmic reticulum (ER) membrane protein which plays a role as a molecular chaperone for the newly synthesized transmembrane proteins. BAP31 is also an important apoptosis regulator for extrinsic apoptosis induction in the ER membrane. Recent studies have shown that BAP31 is also expressed on the surface of embryonic stem cells. However, the function of cell surface BAP31 (csBAP31) still remains unclarified. In an attempt to search for surface markers on tumorspheres, here, we generated monoclonal antibodies (MAbs) against the sphere cells from the non-small cell lung carcinoma cell (NSCLC) line A549. SP1-B7, one of the MAbs, recognized csBAP31 whose expression was further increased on A549 sphere cells, as compared with A549 adherent cells. To investigate the role of csBAP31 in A549 cells, A549 adherent and sphere cells were stained with annexin V, propidium iodide, and SP1-B7. Interestingly, annexin V-high cells showed increased expression of csBAP31 as compared with annexin V-low cells. Caspase-3/7 activity was also increased in csBAP31-high cells as compared with csBAP31-low cells, suggesting that csBAP31-high cells are more sensitive to apoptosis. To further demonstrate the survival of csBAP31-positive A549 cells, csBAP31-positive and -negative A549 cells were sorted and subjected to the clonogenic survival assay. The colony number of csBAP31-positive A549 cells was decreased by approximately 1.7-fold, as compared that of csBAP31-negative A549 cells, suggesting that csBAP31-positve cells are sensitive to cell death indeed. The results suggest that enhanced expression of csBAP31 contributes to poor survival of NSCLC cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Survival , Membrane Proteins/metabolism , Annexin A5/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Humans , Up-Regulation
8.
Sci Rep ; 7(1): 13201, 2017 10 16.
Article in English | MEDLINE | ID: mdl-29038587

ABSTRACT

Circulating tumor cells (CTCs) play a major role in the metastasis and recurrence of hepatocellular carcinoma (HCC). Here, we found that major vault protein (MVP) is expressed on the surface of HCC cells and further induced under stressful environments. MVP knockdown reduces cell proliferation and induces apoptosis in HCC cells. Treatment of HCC cells with anti-MVP antibody (α-MVP) recognizing cell-surface MVP (csMVP) inhibits cell proliferation, migration, and invasion. csMVP-positive HCC cells have a higher clonogenic survival than csMVP-negative HCC cells, and treatment of HCC cells with α-MVP inhibits clonogenic survival, suggesting that csMVP contributes to HCC cell survival, migration, and invasion. The function of csMVP is mediated through mTOR, FAK, ERK and Akt signaling pathways. csMVP-positive CTCs are detected in HCC patients (89.7%) but not in healthy donors, and the number of csMVP-positive CTCs is further increased in patients with metastatic cancers. csMVP is exclusively detectable in CTCs with mesenchymal phenotype or intermediate phenotype with neither epithelial nor mesenchymal markers, suggesting that csMVP-associated survival and metastatic potential harbor CTCs with nonepithelial phenotypes. The results suggest that csMVP promotes cancer progression and serves as a surface marker for mesenchymal and intermediate CTCs in patients with HCC and metastatic cancers.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Apoptosis/genetics , Apoptosis/physiology , Biomarkers, Tumor/genetics , Blotting, Western , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/physiology , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Flow Cytometry , Gene Expression Regulation, Neoplastic/genetics , Hep G2 Cells , Humans , Immunohistochemistry , Immunoprecipitation , Neoplastic Cells, Circulating/metabolism
9.
11.
PLoS One ; 11(12): e0169091, 2016.
Article in English | MEDLINE | ID: mdl-28036384

ABSTRACT

Many studies have shown that the mycoplasmal membrane protein p37 enhances cancer cell migration, invasion, and metastasis. Previously, we generated 6 monoclonal antibodies (MAbs) against the mycoplasmal protein p37 and showed the presence of mycoplasma-infected circulating tumor cells in the blood of hepatocellular carcinoma patients by using CA27, one of the six MAbs. When mycoplasmas were incubated with cancer cells in the presence of CA27, mycoplasma infection was completely inhibited, suggesting that CA27 is a neutralizing antibody inhibiting mycoplasma infection. To examine the neutralizing epitope of CA27, we generated a series of glutathione S-transferase (GST)-fused p37 deletion mutant proteins in which p37 was partly deleted. To express p37-coding sequences in E.coli, mycoplasmal TGA codons were substituted with TGG in the p37 deletion mutant genes. GST-fused p37 deletion mutant proteins were then screened to identify the epitope targeted by CA27. Western blots showed that CA27 bound to the residues 216-246 on the middle part of the p37 protein while it did not bind to the residues 183-219 and 216-240. Fine mapping showed that CA27 was able to bind to the residues 226-246, but its binding activity was relatively weakened as compared to that to the residues 216-246, suggesting that the residues 226-246 is essential for optimal binding activity of CA27. Interestingly, the treatment of the purified GST-tagged epitopes with urea showed that CA27 binding to the epitope was sodium dodecyl sulfate-resistant but urea-sensitive. The same 226-246 residues were also recognized by two other anti-p37 MAbs, suggesting that the epitope is immunodominant. The identification of the novel neutralizing epitope may provide new insight into the interaction between the p37 protein and host receptors.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Epitopes/immunology , Membrane Proteins/immunology , Mycoplasma/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , A549 Cells , Antibodies, Neutralizing/immunology , Carcinoma, Hepatocellular/blood , Cell Line, Tumor , Epitope Mapping , Gene Deletion , Glutathione Transferase/genetics , Humans , Liver Neoplasms/blood , Mycoplasma Infections/immunology , Mycoplasma Infections/microbiology , Recombinant Fusion Proteins/immunology
12.
Int J Stem Cells ; 9(2): 264-270, 2016 Nov 30.
Article in English | MEDLINE | ID: mdl-27871153

ABSTRACT

BACKGROUND: The normal cells derived from human embryonic stem cells (hESCs) are regarded as substitutes for damaged or dysfunctional adult cells. However, tumorigenicity of hESCs remains a major challenge in clinical application of hESC-derived cell transplantation. Previously, we generated monoclonal antibody (MAb) 57-C11 specific to the surface molecule on undifferentiated hESCs. The aim of this study is to prove whether 57-C11-positive hESCs are pluripotent and tumorigenic in immunodeficient mice. METHODS: Undifferentiated hESCs were mixed with retinoic acid (RA)-differentiated hESCs at different ratios prior to 57-C11-mediated separation. To isolate 57-C11-positive hESCs from the mixture, biotinylated 57-C11 and streptavidin-coated magnetic beads were added to the mixture. Unbound 57-C11-negative hESCs were first isolated after applying magnet to the cell mixture, and 57-C11-bound hESCs were then released from the magnetic beads. In order to measure the efficiency of separation, 57-C11-positive or -negative hESCs were counted after isolation. To evaluate the efficiency of teratoma formation in vivo, 57-C11-positive or negative cells were further injected into left and right, respectively, testes of nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. RESULTS: Approximately 77~100% of undifferentiated hESCs were isolated after applying 57-C11-coated magnetic beads to the mixed cell populations. Importantly, teratomas were not observed in NOD/SCID mice after the injection of isolated 57-C11-negative hESCs, whereas teratomas were observed with 57-C11-positive hESCs. CONCLUSION: 57-C11-positive hESCs are pluripotent and tumorigenic. The combination of 57-C11 and magnetic beads will be useful to eliminate remaining undifferentiated hESCs for the safe cell transplantation.

13.
Arterioscler Thromb Vasc Biol ; 36(1): 86-96, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26586661

ABSTRACT

OBJECTIVE: Comprehensive understanding of the mechanisms regulating angiogenesis might provide new strategies for angiogenic therapies for treating diverse physiological and pathological ischemic conditions. The E-twenty six (ETS) factor Ets variant 2 (ETV2; aka Ets-related protein 71) is essential for the formation of hematopoietic and vascular systems. Despite its indispensable function in vessel development, ETV2 role in adult angiogenesis has not yet been addressed. We have therefore investigated the role of ETV2 in vascular regeneration. APPROACH AND RESULTS: We used endothelial Etv2 conditional knockout mice and ischemic injury models to assess the role of ETV2 in vascular regeneration. Although Etv2 expression was not detectable under steady-state conditions, its expression was readily observed in endothelial cells after injury. Mice lacking endothelial Etv2 displayed impaired neovascularization in response to eye injury, wounding, or hindlimb ischemic injury. Lentiviral Etv2 expression in ischemic hindlimbs led to improved recovery of blood perfusion with enhanced vessel formation. After injury, fetal liver kinase 1 (Flk1), aka VEGFR2, expression and neovascularization were significantly upregulated by Etv2, whereas Flk1 expression and vascular endothelial growth factor response were significantly blunted in Etv2-deficient endothelial cells. Conversely, enforced Etv2 expression enhanced vascular endothelial growth factor-mediated endothelial sprouting from embryoid bodies. Lentiviral Flk1 expression rescued angiogenesis defects in endothelial Etv2 conditional knockout mice after hindlimb ischemic injury. Furthermore, Etv2(+/-); Flk1(+/-) double heterozygous mice displayed a more severe hindlimb ischemic injury response compared with Etv2(+/-) or Flk1(+/-) heterozygous mice, revealing an epistatic interaction between ETV2 and FLK1 in vascular regeneration. CONCLUSIONS: Our study demonstrates a novel obligatory role for the ETV2 in postnatal vascular repair and regeneration.


Subject(s)
Angiogenic Proteins/metabolism , Endothelial Cells/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Regeneration , Transcription Factors/metabolism , Angiogenic Proteins/deficiency , Angiogenic Proteins/genetics , Animals , Cells, Cultured , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/physiopathology , Disease Models, Animal , Endothelial Cells/pathology , Gene Expression Regulation , Gene Transfer Techniques , Genetic Vectors , Heterozygote , Hindlimb , Ischemia/genetics , Ischemia/pathology , Ischemia/physiopathology , Ischemia/therapy , Lentivirus/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Phenotype , Recovery of Function , Signal Transduction , Skin/blood supply , Time Factors , Transcription Factors/deficiency , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing
14.
PLoS One ; 10(6): e0130670, 2015.
Article in English | MEDLINE | ID: mdl-26102500

ABSTRACT

When located in the endoplasmic reticulum (ER) membrane, B-cell receptor associated protein 31 (BAP31) is involved in the export of secreted proteins from the ER to the plasma membrane. In a previous study, we generated two monoclonal antibodies (mAbs), 297-D4 and 144-A8, that bound to surface molecules on human embryonic stem cells (hESCs), but not to surface molecules on mouse embryonic stem cells (mESCs). Subsequent studies revealed that the mAbs recognized BAP31 on the surface of hESCs. To investigate the membrane topology of BAP31 on the cell surface, we first examined the epitope specificity of 297-D4 and 144-A8, as well as a polyclonal anti-BAP31 antibody (α-BAP31). We generated a series of GST-fused BAP31 mutant proteins in which BAP31 was serially deleted at the C- terminus. GST-fused BAP31 mutant proteins were then screened to identify the epitopes targeted by the antibodies. Both 297-D4 and 144-A8 recognized C-terminal residues 208-217, while α-BAP31 recognized C-terminal residues 165-246, of BAP31 on hESCs, suggesting that the C-terminal domain of BAP31 is exposed on the cell surface. The polyclonal antibody α-BAP31 bound to mESCs, which confirmed that the C-terminal domain of BAP31 is also exposed on the surface of these cells. Our results show for the first time the novel membrane topology of cell surface-expressed BAP31 as the extracellular exposure of the BAP31 C-terminal domain was not predicted from previous studies.


Subject(s)
Cell Membrane/immunology , Embryonic Stem Cells/immunology , Membrane Proteins/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Antigen-Antibody Reactions , Antigens, Surface/chemistry , Antigens, Surface/immunology , Binding, Competitive , Cell Line , Epitope Mapping , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Protein Translocation Systems , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Sequence Deletion , Sequence Homology, Amino Acid , Species Specificity
15.
Biotechnol J ; 9(7): 915-20, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24616439

ABSTRACT

Human embryonic stem cells (hESCs) are unique cell populations, possessing both unlimited self-renewal capacity and pluripotency, i.e. the potential to give rise to all kinds of specialized cells in the human body. Marker molecules expressed on the surface of hESCs are important for the identification, characterization, and clinical application of hESCs. Compared with conventional genomics- or proteomics-based approaches, generating monoclonal antibody (mAb) libraries against hESCs using alternative methodologies expands the repertoire of mAbs raised against non-protein markers, for example, glycolipid antigens. Additional information about the conformation and post-translational modification of surface molecules can also be obtained. In this article, we review how mAb libraries against hESC surface markers have been developed using whole-cell and decoy immunization strategies.


Subject(s)
Antibodies, Monoclonal/chemistry , Antigens, Surface/chemistry , Embryonic Stem Cells/cytology , Animals , Cell Differentiation , Genomics/methods , Humans , Immunization , Protein Processing, Post-Translational , Proteomics/methods
16.
Biochem Biophys Res Commun ; 446(2): 620-5, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24637212

ABSTRACT

Many studies have shown that persistent infections of bacteria promote carcinogenesis and metastasis. Infectious agents and their products can modulate cancer progression through the induction of host inflammatory and immune responses. The presence of circulating tumor cells (CTCs) is considered as an important indicator in the metastatic cascade. We unintentionally produced a monoclonal antibody (MAb) CA27 against the mycoplasmal p37 protein in mycoplasma-infected cancer cells during the searching process of novel surface markers of CTCs. Mycoplasma-infected cells were enriched by CA27-conjugated magnetic beads in the peripheral blood mononuclear cells in patients with hepatocellular carcinoma (HCC) and analyzed by confocal microscopy with anti-CD45 and CA27 antibodies. CD45-negative and CA27-positive cells were readily detected in three out of seven patients (range 12-30/8.5 ml blood), indicating that they are mycoplasma-infected circulating epithelial cells. CA27-positive cells had larger size than CD45-positive hematological lineage cells, high nuclear to cytoplasmic ratios and irregular nuclear morphology, which identified them as CTCs. The results show for the first time the existence of mycoplasma-infected CTCs in patients with HCC and suggest a possible correlation between mycoplasma infection and the development of cancer metastasis.


Subject(s)
Antigens, Bacterial/blood , Antigens, Tumor-Associated, Carbohydrate/immunology , Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/secondary , Liver Neoplasms/complications , Mycoplasma Infections/pathology , Neoplastic Cells, Circulating/immunology , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Line , Female , Humans , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mycoplasma Infections/blood , Mycoplasma Infections/immunology , Neoplastic Cells, Circulating/pathology
17.
Monoclon Antib Immunodiagn Immunother ; 32(2): 136-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23607350

ABSTRACT

To study cell surface proteins expressed on human embryonic stem cells (hESCs), we generated a panel of monoclonal antibodies (MAbs) against undifferentiated hESCs by a decoy immunization strategy in a previous study. Two of the MAbs, 63-B6 and 246-D7, bound to human pluripotent stem cells but not to human primary cells such as human peripheral blood mononuclear cells and human lung fibroblasts. They did not bind to either mouse embryonic stem cells or mouse embryonic fibroblasts. The two MAbs had similar binding profiles for many various cancer cells, with few exceptions. Expression of antigens recognized by the two MAbs was rapidly decreased during embryoid body formation of hESCs and gradually increased after initial decrease. The MAbs recognized approximately 130 kDa proteins on the surface of hESCs. Cloning and sequence analysis of antibody genes showed that although the MAbs had exactly the same light chain sequences, they had different heavy chain sequences. Taken together, the results suggest that the two MAbs may recognize two different epitopes of the same or different 130 kDa surface proteins involved in regulating the early differentiation of human pluripotent stem cells and cancer cells.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Antigens, Differentiation/immunology , Embryoid Bodies/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal, Murine-Derived/chemistry , Antibodies, Monoclonal, Murine-Derived/genetics , Antibody Specificity , Antigens, Differentiation/metabolism , Cell Differentiation , Cell Line, Tumor , Coculture Techniques , Feeder Cells , Humans , Hybridomas , Immunoglobulin Variable Region/chemistry , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Pluripotent Stem Cells/metabolism , Protein Binding , Sequence Analysis, DNA
18.
Stem Cells ; 31(12): 2647-58, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23495120

ABSTRACT

Self-renewal and pluripotency of human embryonic stem cells (hESCs) are a complex biological process for maintaining hESC stemness. However, the molecular mechanisms underlying these special properties of hESCs are not fully understood. Heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNP A2/B1) is a multifunctional RNA-binding protein whose expression is related to cell proliferation and carcinogenesis. In this study, we found that hnRNP A2/B1 expression was localized to undifferentiated hESCs and decreased upon differentiation of hESCs. hnRNP A2/B1 knockdown reduced the number of alkaline phosphatase-positive colonies in hESCs and led to a decrease in the expression of pluripotency-associated transcription factors OCT4, NANOG, and SOX2, indicating that hnRNP A2/B1 is essential for hESC self-renewal and pluripotency. hnRNP A2/B1 knockdown increased the expression of gene markers associated with the early development of three germ layers, and promoted the process of epithelial-mesenchymal transition, suggesting that hnRNP A2/B1 is required for maintaining the undifferentiated and epithelial phenotypes of hESCs. hnRNP A2/B1 knockdown inhibited hESC proliferation and induced cell cycle arrest in the G0/G1 phase before differentiation via degradation of cyclin D1, cyclin E, and Cdc25A. hnRNP A2/B1 knockdown increased p27 expression and induced phosphorylation of p53 and Chk1, suggesting that hnRNP A2/B1 also regulates the G1/S transition of hESC cell cycle through the control of p27 expression and p53 and Chk1 activity. Analysis of signaling molecules further revealed that hnRNP A2/B1 regulated hESC proliferation in a PI3K/Akt-dependent manner. These findings provide for the first time mechanistic insights into how hnRNP A2/B1 regulates hESC self-renewal and pluripotency.


Subject(s)
Embryonic Stem Cells/cytology , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation/physiology , Cell Growth Processes/physiology , Coculture Techniques , Down-Regulation , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , G1 Phase/physiology , Gene Knockdown Techniques , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Humans , Mice , Pluripotent Stem Cells/metabolism , S Phase/physiology , Transfection
19.
Mol Med Rep ; 5(4): 1023-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22294296

ABSTRACT

The aim of this study was to determine whether the inflammatory milieu and/or hypoxia induces the dedifferentiation of synovial cells into mesenchymal stem-like cells, which may contribute to the tumor-like growth of synovial cells. Expression of mesenchymal stem cell markers (CD24, CD44, CD90, CD106, CD146 and Stro-1) was compared among cultured fibroblast-like synoviocytes (FLSs) from patients with rheumatoid arthritis (RA) or osteoarthritis (OA), bone marrow mesenchymal stem cells (BM MSCs) and normal dermal fibroblasts. After the cells were stimulated with pro-inflammatory cytokines for 3 days under hypoxia or normoxia, the stem cell markers were analyzed by FACS. CD44 and CD90 were expressed constitutively in all four cell types. Only the BM MSCs strongly expressed CD146. The expression of stem cell markers was similar between FLSs from RA and those from OA patients. In addition, the expression levels in FLSs were similar to those in normal dermal fibroblasts. The stimulation of FLSs and dermal fibroblasts with IL-1ß or a mixture of cytokines under hypoxia did not induce a marked change in the expression of stem cell markers. These results indirectly suggest that the pro-inflammatory milieu may be not sufficient to induce the dedifferentiation of FLSs in arthritic joints.


Subject(s)
Cell Differentiation , Cytokines/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Synovial Membrane/cytology , Synovial Membrane/metabolism , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Bone Marrow Cells/cytology , CD146 Antigen/metabolism , Cell Hypoxia , Cells, Cultured , Humans , Hyaluronan Receptors/metabolism , Interleukin-1beta/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Synovial Membrane/drug effects , Thy-1 Antigens/metabolism
20.
Hybridoma (Larchmt) ; 30(4): 401-4, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21851243

ABSTRACT

To investigate cell surface antigens on human embryonic stem cells (hESCs), we generated a panel of monoclonal antibodies (MAbs) against undifferentiated hESCs by a decoy immunization strategy. One of the MAbs, MAb 2-E2, specifically bound to human pluripotent stem cells but not to mouse pluripotent stem cells and mouse embryonic fibroblasts. 2-E2 also bound to human differentiated cells, peripheral blood monocytes, and dermal fibroblasts. 2-E2 antigen expression drastically decreased in retinoic acid-induced differentiated hESCs. However, it gradually increased after initial decrease during embryoid body formation of hESCs. 2-E2 recognized approximately 68 and 27 kDa proteins present on the surface of human pluripotent stem cells. The results suggest that 2-E2-specific surface protein is a novel cell surface protein that plays a role in the early differentiation of human pluripotent stem cells.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , 12E7 Antigen , Animals , Cell Differentiation , Cell Line , Coculture Techniques , Embryonic Stem Cells/cytology , Flow Cytometry , Humans , Leukocytes, Mononuclear/metabolism , Mice , Pluripotent Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...