Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 170
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 147(5): 1146-58, 2011 Nov 23.
Article in English | MEDLINE | ID: mdl-22118468

ABSTRACT

Hematopoietic stem cells (HSCs) reside and self-renew in the bone marrow (BM) niche. Overall, the signaling that regulates stem cell dormancy in the HSC niche remains controversial. Here, we demonstrate that TGF-ß type II receptor-deficient HSCs show low-level Smad activation and impaired long-term repopulating activity, underlining the critical role of TGF-ß/Smad signaling in HSC maintenance. TGF-ß is produced as a latent form by a variety of cells, so we searched for those that express activator molecules for latent TGF-ß. Nonmyelinating Schwann cells in BM proved responsible for activation. These glial cells ensheathed autonomic nerves, expressed HSC niche factor genes, and were in contact with a substantial proportion of HSCs. Autonomic nerve denervation reduced the number of these active TGF-ß-producing cells and led to rapid loss of HSCs from BM. We propose that glial cells are components of a BM niche and maintain HSC hibernation by regulating activation of latent TGF-ß.


Subject(s)
Bone Marrow/physiology , Hematopoietic Stem Cells/cytology , Schwann Cells/cytology , Transforming Growth Factor beta3/metabolism , Animals , Antigens, CD34/metabolism , Hematopoietic Stem Cells/physiology , Humans , Mice , Mice, Inbred C57BL , Neuroglia/metabolism , Schwann Cells/physiology , Sympathectomy
2.
Nature ; 577(7789): 260-265, 2020 01.
Article in English | MEDLINE | ID: mdl-31853061

ABSTRACT

Chronic inflammation is accompanied by recurring cycles of tissue destruction and repair and is associated with an increased risk of cancer1-3. However, how such cycles affect the clonal composition of tissues, particularly in terms of cancer development, remains unknown. Here we show that in patients with ulcerative colitis, the inflamed intestine undergoes widespread remodelling by pervasive clones, many of which are positively selected by acquiring mutations that commonly involve the NFKBIZ, TRAF3IP2, ZC3H12A, PIGR and HNRNPF genes and are implicated in the downregulation of IL-17 and other pro-inflammatory signals. Mutational profiles vary substantially between colitis-associated cancer and non-dysplastic tissues in ulcerative colitis, which indicates that there are distinct mechanisms of positive selection in both tissues. In particular, mutations in NFKBIZ are highly prevalent in the epithelium of patients with ulcerative colitis but rarely found in both sporadic and colitis-associated cancer, indicating that NFKBIZ-mutant cells are selected against during colorectal carcinogenesis. In further support of this negative selection, we found that tumour formation was significantly attenuated in Nfkbiz-mutant mice and cell competition was compromised by disruption of NFKBIZ in human colorectal cancer cells. Our results highlight common and discrete mechanisms of clonal selection in inflammatory tissues, which reveal unexpected cancer vulnerabilities that could potentially be exploited for therapeutics in colorectal cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Colitis, Ulcerative/genetics , Mutation Rate , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colorectal Neoplasms/genetics , Humans , Mice , Signal Transduction
3.
Mol Cell ; 68(3): 626-640.e5, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29107535

ABSTRACT

Eukaryotic cells spend most of their life in interphase of the cell cycle. Understanding the rich diversity of metabolic and genomic regulation that occurs in interphase requires the demarcation of precise phase boundaries in situ. Here, we report the properties of two genetically encoded fluorescence sensors, Fucci(CA) and Fucci(SCA), which enable real-time monitoring of interphase and cell-cycle biology. We re-engineered the Cdt1-based sensor from the original Fucci system to respond to S phase-specific CUL4Ddb1-mediated ubiquitylation alone or in combination with SCFSkp2-mediated ubiquitylation. In cultured cells, Fucci(CA) produced a sharp triple color-distinct separation of G1, S, and G2, while Fucci(SCA) permitted a two-color readout of G1 and S/G2. Fucci(CA) applications included tracking the transient G1 phase of rapidly dividing mouse embryonic stem cells and identifying a window for UV-irradiation damage in S phase. These results show that Fucci(CA) is an essential tool for quantitative studies of interphase cell-cycle regulation.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Cullin Proteins/metabolism , Embryonic Stem Cells/physiology , Fluorescent Dyes/metabolism , Luminescent Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Cullin Proteins/genetics , Embryonic Stem Cells/cytology , Genes, Reporter , HeLa Cells , Humans , Luminescent Proteins/genetics , Mice
4.
Mol Cell ; 66(5): 622-634.e8, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28575658

ABSTRACT

RFWD3 is a recently identified Fanconi anemia protein FANCW whose E3 ligase activity toward RPA is essential in homologous recombination (HR) repair. However, how RPA ubiquitination promotes HR remained unknown. Here, we identified RAD51, the central HR protein, as another target of RFWD3. We show that RFWD3 polyubiquitinates both RPA and RAD51 in vitro and in vivo. Phosphorylation by ATR and ATM kinases is required for this activity in vivo. RFWD3 inhibits persistent mitomycin C (MMC)-induced RAD51 and RPA foci by promoting VCP/p97-mediated protein dynamics and subsequent degradation. Furthermore, MMC-induced chromatin loading of MCM8 and RAD54 is defective in cells with inactivated RFWD3 or expressing a ubiquitination-deficient mutant RAD51. Collectively, our data reveal a mechanism that facilitates timely removal of RPA and RAD51 from DNA damage sites, which is crucial for progression to the late-phase HR and suppression of the FA phenotype.


Subject(s)
Chromatin/enzymology , DNA Damage , DNA/metabolism , Fanconi Anemia/enzymology , Rad51 Recombinase/metabolism , Recombinational DNA Repair , Replication Protein A/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Binding Sites , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromatin/drug effects , Chromatin/genetics , Chromatin/radiation effects , DNA/genetics , Fanconi Anemia/genetics , Humans , Minichromosome Maintenance Proteins/metabolism , Mitomycin/pharmacology , Mutation , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , RNA Interference , Rad51 Recombinase/genetics , Recombinational DNA Repair/drug effects , Recombinational DNA Repair/radiation effects , Replication Protein A/genetics , Transfection , Ubiquitin-Protein Ligases/genetics , Valosin Containing Protein
5.
Cancer Sci ; 114(8): 3259-3269, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37208931

ABSTRACT

Experimental techniques for patient-derived cancer stem-cell organoids/spheroids can be powerful diagnostic tools for personalized chemotherapy. However, establishing their cultures from gastric cancer remains challenging due to low culture efficiency and cumbersome methods. To propagate gastric cancer cells as highly proliferative stem-cell spheroids in vitro, we initially used a similar method to that for colorectal cancer stem cells, which, unfortunately, resulted in a low success rate (25%, 18 of 71 cases). We scrutinized the protocol and found that the unsuccessful cases were largely caused by the paucity of cancer stem cells in the sampled tissues as well as insufficient culture media. To overcome these obstacles, we extensively revised our sample collection protocol and culture conditions. We then investigated the following second cohort and, consequently, achieved a significantly higher success rate (88%, 29 of 33 cases). One of the key improvements included new sampling procedures for tumor tissues from wider and deeper areas of gastric cancer specimens, which allowed securing cancer stem cells more reproducibly. Additionally, we embedded tumor epithelial pieces separately in both Matrigel and collagen type-I as their preference to the extracellular matrix was different depending on the tumors. We also added a low concentration of Wnt ligands to the culture, which helped the growth of occasional Wnt-responsive gastric cancer stem-cell spheroids without allowing proliferation of the normal gastric epithelial stem cells. This newly improved spheroid culture method may facilitate further studies, including personalized drug-sensitivity tests prior to drug therapy.


Subject(s)
Spheroids, Cellular , Stomach Neoplasms , Humans , Spheroids, Cellular/pathology , Stomach Neoplasms/pathology , Neoplastic Stem Cells/pathology
6.
Cell ; 132(3): 487-98, 2008 Feb 08.
Article in English | MEDLINE | ID: mdl-18267078

ABSTRACT

The cell-cycle transition from G1 to S phase has been difficult to visualize. We have harnessed antiphase oscillating proteins that mark cell-cycle transitions in order to develop genetically encoded fluorescent probes for this purpose. These probes effectively label individual G1 phase nuclei red and those in S/G2/M phases green. We were able to generate cultured cells and transgenic mice constitutively expressing the cell-cycle probes, in which every cell nucleus exhibits either red or green fluorescence. We performed time-lapse imaging to explore the spatiotemporal patterns of cell-cycle dynamics during the epithelial-mesenchymal transition of cultured cells, the migration and differentiation of neural progenitors in brain slices, and the development of tumors across blood vessels in live mice. These mice and cell lines will serve as model systems permitting unprecedented spatial and temporal resolution to help us better understand how the cell cycle is coordinated with various biological events.


Subject(s)
Cell Cycle , Cytological Techniques , Animals , COS Cells , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cells, Cultured , Chlorocebus aethiops , Fluorescence , Geminin , HeLa Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal , Molecular Sequence Data , Morphogenesis , Neoplasms/pathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ubiquitination
8.
Mol Cell ; 55(1): 73-84, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24910096

ABSTRACT

Senescence is a state of permanent growth arrest and is a pivotal part of the antitumorigenic barrier in vivo. Although the tumor suppressor activities of p53 and pRb family proteins are essential for the induction of senescence, molecular mechanisms by which these proteins induce senescence are still not clear. Using time-lapse live-cell imaging, we demonstrate here that normal human diploid fibroblasts (HDFs) exposed to various senescence-inducing stimuli undergo a mitosis skip before entry into permanent cell-cycle arrest. This mitosis skip is mediated by both p53-dependent premature activation of APC/C(Cdh1) and pRb family protein-dependent transcriptional suppression of mitotic regulators. Importantly, mitotic skipping is necessary and sufficient for senescence induction. p16 is only required for maintenance of senescence. Analysis of human nevi also suggested the role of mitosis skip in in vivo senescence. Our findings provide decisive evidence for the molecular basis underlying the induction and maintenance of cellular senescence.


Subject(s)
Cellular Senescence , Mitosis/physiology , Cell Cycle Checkpoints , Cyclin-Dependent Kinase Inhibitor p16 , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Models, Biological , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , Time-Lapse Imaging , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/physiology
9.
Proc Natl Acad Sci U S A ; 116(26): 12996-13005, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31182574

ABSTRACT

Cancer stem cell (CSC)-specific markers may be potential therapeutic targets. We previously identified that Dclk1, a tuft cell marker, marks tumor stem cells (TSCs) in mouse intestinal adenomas. Based on the analysis of mouse Dclk1+ tumor cells, we aimed to identify a CSC-specific cell surface marker in human colorectal cancers (hCRCs) and validate the therapeutic effect of targeting it. IL17RB was distinctively expressed by Dclk1+ mouse intestinal tumor cells. Using Il17rb-CreERT2-IRES-EGFP mice, we show that IL17RB marked intestinal TSCs in an IL13-dependent manner. Tuft cell-like cancer cells were detected in a subset of hCRCs. In these hCRCs, lineage-tracing experiments in CRISPR-Cas9-mediated IL17RB-CreERT2 knockin organoids and xenograft tumors revealed that IL17RB marks CSCs that expand independently of IL-13. We observed up-regulation of POU2F3, a master regulator of tuft cell differentiation, and autonomous tuft cell-like cancer cell differentiation in the hCRCs. Furthermore, long-term ablation of IL17RB-expressing CSCs strongly suppressed the tumor growth in vivo. These findings reveal insights into a CSC-specific marker IL17RB in a subset of hCRCs, and preclinically validate IL17RB+ CSCs as a cancer therapeutic target.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Receptors, Interleukin-17/metabolism , Animals , Biomarkers, Tumor/genetics , CRISPR-Cas Systems/genetics , Carcinogenesis , Cell Differentiation , Cell Lineage , Doublecortin-Like Kinases , Gene Knock-In Techniques , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Mice , Mice, Transgenic , Octamer Transcription Factors/metabolism , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/metabolism , Receptors, Interleukin-17/genetics , Spheroids, Cellular , Time-Lapse Imaging , Tumor Cells, Cultured , Up-Regulation , Xenograft Model Antitumor Assays
10.
Proc Natl Acad Sci U S A ; 116(5): 1704-1713, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30635419

ABSTRACT

Inactivating mutations of Arid1a, a subunit of the Switch/sucrose nonfermentable chromatin remodeling complex, have been reported in multiple human cancers. Intestinal deletion of Arid1a has been reported to induce colorectal cancer in mice; however, its functional role in intestinal homeostasis remains unclear. We investigated the functional role of Arid1a in intestinal homeostasis in mice. We found that intestinal deletion of Arid1a results in loss of intestinal stem cells (ISCs), decreased Paneth and goblet cells, disorganized crypt-villous structures, and increased apoptosis in adult mice. Spheroids did not develop from intestinal epithelial cells deficient for Arid1a Lineage-tracing experiments revealed that Arid1a deletion in Lgr5+ ISCs leads to impaired self-renewal of Lgr5+ ISCs but does not perturb intestinal homeostasis. The Wnt signaling pathway, including Wnt agonists, receptors, and target genes, was strikingly down-regulated in Arid1a-deficient intestines. We found that Arid1a directly binds to the Sox9 promoter to support its expression. Remarkably, overexpression of Sox9 in intestinal epithelial cells abrogated the above phenotypes, although Sox9 overexpression in intestinal epithelial cells did not restore the expression levels of Wnt agonist and receptor genes. Furthermore, Sox9 overexpression permitted development of spheroids from Arid1a-deficient intestinal epithelial cells. In addition, deletion of Arid1a concomitant with Sox9 overexpression in Lgr5+ ISCs restores self-renewal in Arid1a-deleted Lgr5+ ISCs. These results indicate that Arid1a is indispensable for the maintenance of ISCs and intestinal homeostasis in mice. Mechanistically, this is mainly mediated by Sox9. Our data provide insights into the molecular mechanisms underlying maintenance of ISCs and intestinal homeostasis.


Subject(s)
DNA-Binding Proteins/metabolism , Intestinal Mucosa/metabolism , Nuclear Proteins/metabolism , SOX9 Transcription Factor/metabolism , Stem Cells/metabolism , Animals , Epithelial Cells/metabolism , Homeostasis/physiology , Intestines/physiology , Mice , Promoter Regions, Genetic/physiology , Transcription Factors , Wnt Signaling Pathway/physiology
11.
EMBO J ; 36(1): 5-24, 2017 01 04.
Article in English | MEDLINE | ID: mdl-27797821

ABSTRACT

Adaptive cellular responses are often required during wound repair. Following disruption of the intestinal epithelium, wound-associated epithelial (WAE) cells form the initial barrier over the wound. Our goal was to determine the critical factor that promotes WAE cell differentiation. Using an adaptation of our in vitro primary epithelial cell culture system, we found that prostaglandin E2 (PGE2) signaling through one of its receptors, Ptger4, was sufficient to drive a differentiation state morphologically and transcriptionally similar to in vivo WAE cells. WAE cell differentiation was a permanent state and dominant over enterocyte differentiation in plasticity experiments. WAE cell differentiation was triggered by nuclear ß-catenin signaling independent of canonical Wnt signaling. Creation of WAE cells via the PGE2-Ptger4 pathway was required in vivo, as mice with loss of Ptger4 in the intestinal epithelium did not produce WAE cells and exhibited impaired wound repair. Our results demonstrate a mechanism by which WAE cells are formed by PGE2 and suggest a process of adaptive cellular reprogramming of the intestinal epithelium that occurs to ensure proper repair to injury.


Subject(s)
Cell Differentiation , Dinoprostone/metabolism , Epithelial Cells/physiology , Intestinal Mucosa/injuries , Intestinal Mucosa/physiology , Wound Healing , Animals , Mice , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Signal Transduction
12.
Neurosurg Rev ; 44(1): 29-49, 2021 Feb.
Article in English | MEDLINE | ID: mdl-31781985

ABSTRACT

Malignant glioma, which is characterized by diffuse infiltration into the normal brain parenchyma, is the most aggressive primary brain tumor with dismal prognosis. Over the past 40 years, the median survival has only slightly improved. Therefore, new therapeutic modalities must be developed. In the 1990s, suicide gene therapy began attracting attention for the treatment of malignant glioma. Some clinical trials used a viral vector for suicide gene transduction; however, it was found that viral vectors cannot cover the large invaded area of glioma cells. Interest in this therapy was recently revived because some types of stem cells possess a tumor-tropic migratory capacity, which can be used as cellular delivery vehicles. Immortalized, clonal neural stem cell (NSC) line has been used for patients with recurrent high-grade glioma, which showed safety and efficacy. Embryonic and induced pluripotent stem cells may be considered as sources of NSC because NSC is difficult to harvest, and ethical issues have been raised. Mesenchymal stem cells are alternative candidates for cellular vehicle and are easily harvested from the bone marrow. In addition, a new type of nonlytic, amphotropic retroviral replicating vector encoding suicide gene has shown efficacy in patients with recurrent high-grade glioma in a clinical trial. This replicating viral capacity is another possible candidate as delivery vehicle to tackle gliomas. Herein, we review the concept of suicide gene therapy, as well as recent progress in preclinical and clinical studies in this field.


Subject(s)
Brain Neoplasms/therapy , Genes, Transgenic, Suicide/genetics , Genetic Therapy/methods , Glioma/therapy , Clinical Trials as Topic , Humans
13.
Genes Cells ; 23(7): 580-589, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29749672

ABSTRACT

Jellyfish green fluorescent protein (GFP) and firefly luciferase can serve as versatile tracking markers for identification and quantification of transplanted cancer cells in vivo. However, immune reactions against these markers can hamper the formation of syngraft tumors and metastasis that follows. Here, we report two transgenic (Tg) mouse lines that express nonfunctional mutant marker proteins, namely modified firefly luciferase (Luc2) or enhanced GFP (EGFP). These mice, named as Tg-mLuc2 and Tg-mEGFP, turned out to be immunologically tolerant to the respective tracking markers and thus efficiently accepted syngeneic cancer cells expressing the active forms of the markers. We then injected intrarectally the F1 hybrid Tg mice (BALB/c × C57BL/6J) with Colon-26 (C26) colon cancer cells that originated from a BALB/c mouse. Even when C26 cells expressed active Luc2 or EGFP, they formed primary tumors in the Tg mice with only 104 cells per mouse compared with more than 106 cells required in the nontransgenic BALB/c hosts. Furthermore, we detected metastatic foci of C26 cells in the liver and lungs of the Tg mice by tracking the specific reporter activities. These results show the usefulness of the Tg mouse lines as recipients for transplantation experiments with the non-self tracking marker-expressing cells.


Subject(s)
Isografts/metabolism , Neoplasm Transplantation/methods , Animals , Green Fluorescent Proteins , Luciferases , Luminescent Proteins , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic/physiology , Neoplasms
14.
Biol Reprod ; 100(6): 1440-1452, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30869744

ABSTRACT

Nonhuman primates (NHPs) are considered to be the most valuable models for human transgenic (Tg) research into disease because human pathology is more closely recapitulated in NHPs than rodents. Previous studies have reported the generation of Tg NHPs that ubiquitously overexpress a transgene using various promoters, but it is not yet clear which promoter is most suitable for the generation of NHPs overexpressing a transgene ubiquitously and persistently in various tissues. To clarify this issue, we evaluated four putative ubiquitous promoters, cytomegalovirus (CMV) immediate-early enhancer and chicken beta-actin (CAG), elongation factor 1α (EF1α), ubiquitin C (UbC), and CMV, using an in vitro differentiation system of cynomolgus monkey embryonic stem cells (ESCs). While the EF1α promoter drove Tg expression more strongly than the other promoters in undifferentiated pluripotent ESCs, the CAG promoter was more effective in differentiated cells such as embryoid bodies and ESC-derived neurons. When the CAG and EF1α promoters were used to generate green fluorescent protein (GFP)-expressing Tg monkeys, the CAG promoter drove GFP expression in skin and hematopoietic tissues more strongly than in ΕF1α-GFP Tg monkeys. Notably, the EF1α promoter underwent more silencing in both ESCs and Tg monkeys. Thus, the CAG promoter appears to be the most suitable for ubiquitous and stable expression of transgenes in the differentiated tissues of Tg cynomolgus monkeys and appropriate for the establishment of human disease models.


Subject(s)
Animals, Genetically Modified , Genetic Vectors , Macaca fascicularis/genetics , Promoter Regions, Genetic , Transgenes , Actins/genetics , Animals , Antigens, Viral/genetics , Cells, Cultured , Chickens/genetics , Cloning, Organism/methods , Cloning, Organism/standards , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Enhancer Elements, Genetic/genetics , Female , Gene Transfer Techniques/standards , Genetic Vectors/genetics , Immediate-Early Proteins/genetics , Male , Mice , Peptide Elongation Factor 1/genetics
15.
Int J Mol Sci ; 20(4)2019 Feb 14.
Article in English | MEDLINE | ID: mdl-30769780

ABSTRACT

Human induced pluripotent stem cells (iPSCs) hold enormous promise for regenerative medicine. The major safety concern is the tumorigenicity of transplanted cells derived from iPSCs. A potential solution would be to introduce a suicide gene into iPSCs as a safety switch. The herpes simplex virus type 1 thymidine kinase (HSV-TK) gene, in combination with ganciclovir, is the most widely used enzyme/prodrug suicide system from basic research to clinical applications. In the present study, we attempted to establish human iPSCs that stably expressed HSV-TK with either lentiviral vectors or CRISPR/Cas9-mediated genome editing. However, this task was difficult to achieve, because high-level and/or constitutive expression of HSV-TK resulted in the induction of cell death or silencing of HSV-TK expression. A nucleotide metabolism analysis suggested that excessive accumulation of thymidine triphosphate, caused by HSV-TK expression, resulted in an imbalance in the dNTP pools. This unbalanced state led to DNA synthesis inhibition and cell death in a process similar to a "thymidine block", but more severe. We also demonstrated that the Tet-inducible system was a feasible solution for overcoming the cytotoxicity of HSV-TK expression. Our results provided a warning against using the HSV-TK gene in human iPSCs, particularly in clinical applications.


Subject(s)
Genetic Therapy , Induced Pluripotent Stem Cells/enzymology , Simplexvirus/enzymology , Thymidine Kinase/genetics , Apoptosis/genetics , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Ganciclovir/pharmacology , Gene Editing , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Viral/genetics , Genes, Transgenic, Suicide/genetics , Genetic Vectors/therapeutic use , Humans , Induced Pluripotent Stem Cells/transplantation , Lentivirus/genetics , Nucleotides/biosynthesis , Nucleotides/genetics , Simplexvirus/genetics
16.
Stem Cells ; 35(5): 1316-1327, 2017 05.
Article in English | MEDLINE | ID: mdl-28142229

ABSTRACT

Although human induced pluripotent stem cell (hiPSC) derivatives are considered promising cellular resources for regenerative medicine, their tumorigenicity potentially limits their clinical application in hiPSC technologies. We previously demonstrated that oncogenic hiPSC-derived neural stem/progenitor cells (hiPSC-NS/PCs) produced tumor-like tissues that were distinct from teratomas. To gain insight into the mechanisms underlying the regulation of tumorigenicity in hiPSC-NS/PCs, we performed an integrated analysis using the Infinium HumanMethylation450 BeadChip array and the HumanHT-12 v4.0 Expression BeadChip array to compare the comprehensive DNA methylation and gene expression profiles of tumorigenic hiPSC-NS/PCs (253G1-NS/PCs) and non-tumorigenic cells (201B7-NS/PCs). Although the DNA methylation profiles of 253G1-hiPSCs and 201B7-hiPSCs were similar regardless of passage number, the methylation status of the global DNA methylation profiles of 253G1-NS/PCs and 201B7-NS/PCs differed; the genomic regions surrounding the transcriptional start site of the CAT and PSMD5 genes were hypermethylated in 253G1-NS/PCs but not in 201B7-NS/PCs. Interestingly, the aberrant DNA methylation profile was more pronounced in 253G1-NS/PCs that had been passaged more than 15 times. In addition, we identified aberrations in DNA methylation at the RBP1 gene locus; the DNA methylation frequency in RBP1 changed as 253G1-NS/PCs were sequentially passaged. These results indicate that different NS/PC clones have different DNA methylomes and that DNA methylation patterns are unstable as cells are passaged. Therefore, DNA methylation profiles should be included in the criteria used to evaluate the tumorigenicity of hiPSC-NS/PCs in the clinical setting. Stem Cells 2017;35:1316-1327.


Subject(s)
Carcinogenesis/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Genome, Human , Induced Pluripotent Stem Cells/pathology , Neural Stem Cells/pathology , Biomarkers, Tumor/genetics , Carcinogenesis/pathology , Cell Proliferation/genetics , Gene Expression Profiling , Genes, Tumor Suppressor , Genetic Association Studies , Humans , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Neural Stem Cells/metabolism
17.
Stem Cells ; 35(10): 2115-2128, 2017 10.
Article in English | MEDLINE | ID: mdl-28782268

ABSTRACT

Reprogramming of cancer cells into induced pluripotent stem cells (iPSCs) is a compelling idea for inhibiting oncogenesis, especially through modulation of homeobox proteins in this reprogramming process. We examined the role of various long noncoding RNAs (lncRNAs)-homeobox protein HOXA13 axis on the switching of the oncogenic function of bone morphogenetic protein 7 (BMP7), which is significantly lost in the gastric cancer cell derived iPS-like cells (iPSLCs). BMP7 promoter activation occurred through the corecruitment of HOXA13, mixed-lineage leukemia 1 lysine N-methyltransferase, WD repeat-containing protein 5, and lncRNA HoxA transcript at the distal tip (HOTTIP) to commit the epigenetic changes to the trimethylation of lysine 4 on histone H3 in cancer cells. By contrast, HOXA13 inhibited BMP7 expression in iPSLCs via the corecruitment of HOXA13, enhancer of zeste homolog 2, Jumonji and AT rich interactive domain 2, and lncRNA HoxA transcript antisense RNA (HOTAIR) to various cis-element of the BMP7 promoter. Knockdown experiments demonstrated that HOTTIP contributed positively, but HOTAIR regulated negatively to HOXA13-mediated BMP7 expression in cancer cells and iPSLCs, respectively. These findings indicate that the recruitment of HOXA13-HOTTIP and HOXA13-HOTAIR to different sites in the BMP7 promoter is crucial for the oncogenic fate of human gastric cells. Reprogramming with octamer-binding protein 4 and Jun dimerization protein 2 can inhibit tumorigenesis by switching off BMP7. Stem Cells 2017;35:2115-2128.


Subject(s)
Cellular Reprogramming Techniques/methods , Homeodomain Proteins/genetics , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Cell Line, Tumor , Cell Proliferation , Homeodomain Proteins/metabolism , Humans , Promoter Regions, Genetic , RNA, Long Noncoding/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
18.
Nucleic Acids Res ; 44(22): 10758-10771, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27694619

ABSTRACT

The FANCI-FANCD2 (I-D) complex is considered to work with RAD51 to protect the damaged DNA in the stalled replication fork. However, the means by which this DNA protection is accomplished have remained elusive. In the present study, we found that the I-D complex directly binds to RAD51, and stabilizes the RAD51-DNA filament. Unexpectedly, the DNA binding activity of FANCI, but not FANCD2, is explicitly required for the I-D complex-mediated RAD51-DNA filament stabilization. The RAD51 filament stabilized by the I-D complex actually protects the DNA end from nucleolytic degradation by an FA-associated nuclease, FAN1. This DNA end protection is not observed with the RAD51 mutant from FANCR patient cells. These results clearly answer the currently enigmatic question of how RAD51 functions with the I-D complex to prevent genomic instability at the stalled replication fork.


Subject(s)
Avian Proteins/physiology , DNA/genetics , Fanconi Anemia Complementation Group D2 Protein/physiology , Rad51 Recombinase/physiology , Amino Acid Sequence , Animals , Cell Line, Tumor , Chickens , Conserved Sequence , DNA/metabolism , DNA Damage , DNA Repair , DNA Replication , Genomic Instability , Humans , Protein Binding , Protein Stability , Ubiquitination
19.
Mol Cancer ; 16(1): 140, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28830458

ABSTRACT

BACKGROUND: Glioma stem cells (GSCs) are a major cause of the frequent relapse observed in glioma, due to their high drug resistance and their differentiation potential. Therefore, understanding the molecular mechanisms governing the 'cancer stemness' of GSCs will be particularly important for improving the prognosis of glioma patients. METHODS: We previously established cancerous neural stem cells (CNSCs) from immortalized human neural stem cells (F3 cells), using the H-Ras oncogene. In this study, we utilized the EGFRviii mutation, which frequently occurs in brain cancers, to establish another CNSC line (F3.EGFRviii), and characterized its stemness under spheroid culture. RESULTS: The F3.EGFRviii cell line was highly tumorigenic in vitro and showed high ERK1/2 activity as well as expression of a variety of genes associated with cancer stemness, such as SOX2 and NANOG, under spheroid culture conditions. Through meta-analysis, PCR super-array, and subsequent biochemical assays, the induction of MEK partner-1 (MP1, encoded by the LAMTOR3 gene) was shown to play an important role in maintaining ERK1/2 activity during the acquisition of cancer stemness under spheroid culture conditions. High expression of this gene was also closely associated with poor prognosis in brain cancer. CONCLUSION: These data suggest that MP1 contributes to cancer stemness in EGFRviii-expressing glioma cells by driving ERK activity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , ErbB Receptors/metabolism , MAP Kinase Signaling System/physiology , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Prognosis
20.
EMBO J ; 32(24): 3130-44, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24185898

ABSTRACT

Delivery of granule contents to epithelial surfaces by secretory cells is a critical physiologic process. In the intestine, goblet cells secrete mucus that is required for homeostasis. Autophagy proteins are required for secretion in some cases, though the mechanism and cell biological basis for this requirement remain unknown. We found that in colonic goblet cells, proteins involved in initiation and elongation of autophagosomes were required for efficient mucus secretion. The autophagy protein LC3 localized to intracellular multi-vesicular vacuoles that were consistent with a fusion of autophagosomes and endosomes. Using cultured intestinal epithelial cells, we found that NADPH oxidases localized to and enhanced the formation of these LC3-positive vacuoles. Both autophagy proteins and endosome formation were required for maximal production of reactive oxygen species (ROS) derived from NADPH oxidases. Importantly, generation of ROS was critical to control mucin granule accumulation in colonic goblet cells. Thus, autophagy proteins can control secretory function through ROS, which is in part generated by LC3-positive vacuole-associated NADPH oxidases. These findings provide a novel mechanism by which autophagy proteins can control secretion.


Subject(s)
Autophagy , Goblet Cells/metabolism , Microtubule-Associated Proteins/metabolism , Reactive Oxygen Species/metabolism , Animals , Autophagy-Related Protein 5 , Cells, Cultured , Colon/cytology , Endocytosis , Epithelial Cells/metabolism , Goblet Cells/cytology , Goblet Cells/physiology , Mice , Mice, Mutant Strains , Microtubule-Associated Proteins/genetics , Mucins/metabolism , Mutation , NADPH Oxidases/metabolism , Phagosomes/metabolism , Vesicular Transport Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL