55 results on '"Pengcheng Bu"'
Search Results
2. Supplementary Figure 3 from NOTCH Signaling Regulates Asymmetric Cell Fate of Fast- and Slow-Cycling Colon Cancer–Initiating Cells
- Author
-
Xiling Shen, Steven M. Lipkin, Leonard Augenlicht, Jeff Milsom, Nicole Panarelli, Kai-Yuan Chen, Kuei-Ling Tung, Elaine Bich Than, Pengcheng Bu, Jewell Walters, and Tara Srinivasan
- Abstract
MYC knockdown in CCIC-1 line.
- Published
- 2023
3. Supplementary Movie 1 from NOTCH Signaling Regulates Asymmetric Cell Fate of Fast- and Slow-Cycling Colon Cancer–Initiating Cells
- Author
-
Xiling Shen, Steven M. Lipkin, Leonard Augenlicht, Jeff Milsom, Nicole Panarelli, Kai-Yuan Chen, Kuei-Ling Tung, Elaine Bich Than, Pengcheng Bu, Jewell Walters, and Tara Srinivasan
- Abstract
3D reconstruction of LGR5+/BMI1+ CCIC-1 asymmetric daughter cell pair
- Published
- 2023
4. Supplementary Figure 2 from NOTCH Signaling Regulates Asymmetric Cell Fate of Fast- and Slow-Cycling Colon Cancer–Initiating Cells
- Author
-
Xiling Shen, Steven M. Lipkin, Leonard Augenlicht, Jeff Milsom, Nicole Panarelli, Kai-Yuan Chen, Kuei-Ling Tung, Elaine Bich Than, Pengcheng Bu, Jewell Walters, and Tara Srinivasan
- Abstract
Asymmetry of BMI1+/LGR5+ CCIC-1 pairs.
- Published
- 2023
5. Supplementary Figure 1 from NOTCH Signaling Regulates Asymmetric Cell Fate of Fast- and Slow-Cycling Colon Cancer–Initiating Cells
- Author
-
Xiling Shen, Steven M. Lipkin, Leonard Augenlicht, Jeff Milsom, Nicole Panarelli, Kai-Yuan Chen, Kuei-Ling Tung, Elaine Bich Than, Pengcheng Bu, Jewell Walters, and Tara Srinivasan
- Abstract
Characterization of BMI1+/LGR5+ CCIC-1 asymmetric daughter cell pairs.
- Published
- 2023
6. Data from NOTCH Signaling Regulates Asymmetric Cell Fate of Fast- and Slow-Cycling Colon Cancer–Initiating Cells
- Author
-
Xiling Shen, Steven M. Lipkin, Leonard Augenlicht, Jeff Milsom, Nicole Panarelli, Kai-Yuan Chen, Kuei-Ling Tung, Elaine Bich Than, Pengcheng Bu, Jewell Walters, and Tara Srinivasan
- Abstract
Colorectal cancer cells with stem-like properties, referred to as colon cancer–initiating cells (CCIC), have high tumorigenic potential. While CCIC can differentiate to promote cellular heterogeneity, it remains unclear whether CCIC within a tumor contain distinct subpopulations. Here, we describe the co-existence of fast- and slow-cycling CCIC, which can undergo asymmetric division to generate each other, highlighting CCIC plasticity and interconvertibility. Fast-cycling CCIC express markers, such as LGR5 and CD133, rely on MYC for their proliferation, whereas slow-cycling CCIC express markers, such as BMI1 and hTERT, are independent of MYC. NOTCH signaling promotes asymmetric cell fate, regulating the balance between these two populations. Overall, our results illuminate the basis for CCIC heterogeneity and plasticity by defining a direct interconversion mechanism between slow- and fast-cycling CCIC. Cancer Res; 76(11); 3411–21. ©2016 AACR.
- Published
- 2023
7. Supplementary Figure 5 from NOTCH Signaling Regulates Asymmetric Cell Fate of Fast- and Slow-Cycling Colon Cancer–Initiating Cells
- Author
-
Xiling Shen, Steven M. Lipkin, Leonard Augenlicht, Jeff Milsom, Nicole Panarelli, Kai-Yuan Chen, Kuei-Ling Tung, Elaine Bich Than, Pengcheng Bu, Jewell Walters, and Tara Srinivasan
- Abstract
NOTCH modulation in CCIC-1 line.
- Published
- 2023
8. Supplementary Figure Legends from NOTCH Signaling Regulates Asymmetric Cell Fate of Fast- and Slow-Cycling Colon Cancer–Initiating Cells
- Author
-
Xiling Shen, Steven M. Lipkin, Leonard Augenlicht, Jeff Milsom, Nicole Panarelli, Kai-Yuan Chen, Kuei-Ling Tung, Elaine Bich Than, Pengcheng Bu, Jewell Walters, and Tara Srinivasan
- Abstract
Contains Supplementary Figure legends for Supplementary Figures 1-6.
- Published
- 2023
9. Supplementary Methods from NOTCH Signaling Regulates Asymmetric Cell Fate of Fast- and Slow-Cycling Colon Cancer–Initiating Cells
- Author
-
Xiling Shen, Steven M. Lipkin, Leonard Augenlicht, Jeff Milsom, Nicole Panarelli, Kai-Yuan Chen, Kuei-Ling Tung, Elaine Bich Than, Pengcheng Bu, Jewell Walters, and Tara Srinivasan
- Abstract
Contains information about antibodies used for immunofluorescence and Western blotting and primers for RT-PCR.
- Published
- 2023
10. High Extinction Ratio 4 × 2 Encoder Based on Electro-Optical Graphene Plasma Structure
- Author
-
Aijun Zhu, Pengcheng Bu, Cong Hu, Junhao Niu, and Rabi Mahapatra
- Subjects
Radiology, Nuclear Medicine and imaging ,Instrumentation ,Atomic and Molecular Physics, and Optics - Abstract
In this paper, a plasmonic electro-optical encoder based on graphene at THz frequency is proposed. The surface plasmon polaritons (SPPs) in the graphene–insulator–metal structure are excited by an incident TM wave with a wavelength of 9.3 μm. Graphene plasma waveguides have extremely high confinement, relatively low losses, and high tunability. The switching mechanism is based on the application of an external voltage to locally change the chemical potential of the graphene for encoding. Setting the chemical potential to 1 eV allows SPPs to propagate while lowering the chemical potential to 0.1 eV prevents the SPPs from propagating. A 4 × 2 encoder with a minimum encoding extinction ratio (ER) of 37 dB, a maximum modulation depth (MD) of 99.99%, and a structure area of 0.8 μm2 is proposed based on the design rules and simulations using the finite-difference time-domain (FDTD) method. In terms of the obtained results, the proposed structure can be used in optical integrated circuits.
- Published
- 2023
11. Generation of an orthotopic mouse model to study colorectal cancer metastasis
- Author
-
Liwen Zhang and Pengcheng Bu
- Subjects
Oncology ,medicine.medical_specialty ,Science (General) ,Colorectal cancer ,General Biochemistry, Genetics and Molecular Biology ,Metastasis ,Q1-390 ,Mice ,Model Organisms ,Internal medicine ,Protocol ,Medicine ,Animals ,Humans ,Neoplasm Metastasis ,Cancer ,General Immunology and Microbiology ,business.industry ,Mechanism (biology) ,General Neuroscience ,Neoplasms, Experimental ,medicine.disease ,HCT116 Cells ,digestive system diseases ,Colonic Neoplasms ,business - Abstract
Summary This protocol describes the generation of a mouse colorectal cancer (CRC) model to study the mechanism of CRC growth and metastasis. Compared to existing protocols, this protocol is mainly to improve the incidence of metastasis. The major advantage of this model is that it mimics the process of clinical CRC metastasis. Thus, it can be used to study different stages of CRC progression and to identify molecular markers or therapeutic targets. The limitation is the difficulty of performing the operation. For complete details on the use and execution of this protocol, please refer to Zhang et al. (2021)., Graphical abstract, Highlights • CRC orthotopic mouse model recapitulates human CRC progression including metastasis • CRC orthotopic mouse model generates paired primary CRC tumor and metastases • CRC orthotopic mouse model is a useful tool to study CRC tumor microenvironment, This protocol describes the generation of a mouse colorectal cancer (CRC) model to study the mechanism of CRC growth and metastasis. Compared to existing protocols, this protocol is mainly to improve the incidence of metastasis. The major advantage of this model is that it mimics the process of clinical CRC metastasis. Thus, it can be used to study different stages of CRC progression and to identify molecular markers or therapeutic targets. The limitation is the difficulty of performing the operation.
- Published
- 2021
12. The two sides of creatine in cancer
- Author
-
Liwen Zhang and Pengcheng Bu
- Subjects
Mammals ,Mice ,Phosphocreatine ,Animals ,Humans ,Breast Neoplasms ,Female ,Cell Biology ,Creatine - Abstract
Creatine is a nitrogen-containing organic acid naturally existing in mammals. It can be converted into phosphocreatine to provide energy for muscle and nerve tissues. Creatine and its analog, cyclocreatine, have been considered cancer suppressive metabolites due to their effects on suppression of subcutaneous cancer growth. Recently, emerging studies have demonstrated the promoting effect of creatine on cancer metastasis. Orthotopic mouse models revealed that creatine promoted invasion and metastasis of pancreatic cancer, colorectal cancer, and breast cancer. Thus, creatine possesses considerably complicated roles in cancer progression. In this review, we systematically summarized the role of creatine in tumor progression, which will call to caution when considering creatine supplementation to clinically treat cancer patients.
- Published
- 2021
13. CD146 is a Novel ANGPTL2 Receptor that Promotes Obesity by Manipulating Lipid Metabolism and Energy Expenditure
- Author
-
Zhenzhen Wu, S.R. Yang, Xuehui Chen, Junfeng Du, Huiyun Cai, Yongting Luo, Xiyun Yan, Gaoqi Ye, Hongxia Duan, Yan Cui, Gang Chen, Guizhi Shi, Pingsheng Liu, Jing Feng, Liwen Zhang, Hongjie Zhang, Zheng Liu, Yiyi Luo, Jingyu Liu, Pengcheng Bu, Lei Sun, Taotao Wei, and Hongwei Sun
- Subjects
obesity ,General Chemical Engineering ,white adipocyte ,General Physics and Astronomy ,Medicine (miscellaneous) ,Adipose tissue ,Inflammation ,02 engineering and technology ,010402 general chemistry ,CREB ,01 natural sciences ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Cell surface receptor ,ANGPTL2 ,energy expenditure ,medicine ,General Materials Science ,Receptor ,lcsh:Science ,Full Paper ,biology ,Chemistry ,General Engineering ,Lipid metabolism ,Full Papers ,021001 nanoscience & nanotechnology ,0104 chemical sciences ,Cell biology ,Adipogenesis ,CD146 ,biology.protein ,lcsh:Q ,medicine.symptom ,0210 nano-technology ,brown adipocyte - Abstract
Obesity and its related complications pose an increasing threat to human health; however, targetable obesity‐related membrane receptors are not yet elucidated. Here, the membrane receptor CD146 is demonstrated to play an essential role in obesity. In particular, CD146 acts as a new adipose receptor for angiopoietin‐like protein 2 (ANGPTL2), which is thought to act on endothelial cells to activate adipose inflammation. ANGPTL2 binds to CD146 to activate cAMP response element‐binding protein (CREB), which then upregulates CD146 during adipogenesis and adipose inflammation. CD146 is present in preadipocytes and mature adipocytes, where it is mediated by its ligands ANGPTL2 and galectin‐1. In preadipocytes, CD146 ablation suppresses adipogenesis, whereas the loss of CD146 in mature adipocytes suppresses lipid accumulation and enhances energy expenditure. Moreover, anti‐CD146 antibodies inhibit obesity by disrupting the interactions between CD146 and its ligands. Together, these findings demonstrate that ANGPTL2 directly affects adipocytes via CD146 to promote obesity, suggesting that CD146 can be a potential target for treating obesity., The membrane receptor CD146 is a novel ANGPTL2 receptor that promotes obesity and insulin resistance. By interacting with ANGPTL2 and galectin‐1, CD146 enhances adipogenesis and lipogenesis while suppressing FAO and BAT thermogenesis. Furthermore, the disruption of the interaction between CD146 and its ligands by knocking out CD146 or using anti‐CD146 antibodies may prevent obesity and its related complications.
- Published
- 2021
14. Chromatin Remodeling of Colorectal Cancer Liver Metastasis is Mediated by an HGF‐PU.1‐DPP4 Axis
- Author
-
Wei Li, Anders Dohlman, Jorge Prado Balcazar, Liwen Zhang, Gregory E. Crawford, Kun Xiang, Marcos Negrete, Xiling Shen, Lihua Wang, Purushothama Rao Tata, Zhenzhen Wu, David S. Hsu, Yi Wang, Tianyi Chen, Scott Kopetz, Guizhi Shi, Victor Moreno, Ergang Wang, Emina H. Huang, Charles A. Gersbach, John Paul Shen, Pengcheng Bu, Robert Mines, Qiang Huang, Yoshihiko Kobayashi, Yujie Fu, and Kai-Yuan Chen
- Subjects
Dipeptidyl Peptidase 4 ,General Chemical Engineering ,hepatic growth factor (HGF) ,Science ,General Physics and Astronomy ,Medicine (miscellaneous) ,colorectal cancer ,DPP4 ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Chromatin remodeling ,Epigenesis, Genetic ,Metastasis ,chromatin remodeling ,Mice ,Metàstasi ,Càncer colorectal ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Gene silencing ,metastasis ,General Materials Science ,Epigenetics ,Promoter Regions, Genetic ,Research Articles ,Mice, Inbred BALB C ,biology ,epigenetics ,Hepatocyte Growth Factor ,Liver Neoplasms ,PU.1 ,Pioneer factor ,General Engineering ,Cancer ,Chromatin Assembly and Disassembly ,medicine.disease ,Colorectal cancer ,Chromatin ,Gene Expression Regulation, Neoplastic ,Histone ,Trans-Activators ,biology.protein ,Cancer research ,Colorectal Neoplasms ,Research Article - Abstract
Colorectal cancer (CRC) metastasizes mainly to the liver, which accounts for the majority of CRC‐related deaths. Here it is shown that metastatic cells undergo specific chromatin remodeling in the liver. Hepatic growth factor (HGF) induces phosphorylation of PU.1, a pioneer factor, which in turn binds and opens chromatin regions of downstream effector genes. PU.1 increases histone acetylation at the DPP4 locus. Precise epigenetic silencing by CRISPR/dCas9KRAB or CRISPR/dCas9HDAC revealed that individual PU.1‐remodeled regulatory elements collectively modulate DPP4 expression and liver metastasis growth. Genetic silencing or pharmacological inhibition of each factor along this chromatin remodeling axis strongly suppressed liver metastasis. Therefore, microenvironment‐induced epimutation is an important mechanism for metastatic tumor cells to grow in their new niche. This study presents a potential strategy to target chromatin remodeling in metastatic cancer and the promise of repurposing drugs to treat metastasis., Metastasis of colorectal cancer to the liver is a leading cause of cancer‐related death. This work shows that metastatic colorectal cancer cells undergo chromatin remodeling in the liver via an HGF‐PU.1‐DPP4 axis. Pharmacological or genetic inhibition of this remodeling axis can suppress liver metastasis and prolong survival.
- Published
- 2021
15. Non-coding RNA in cancer
- Author
-
Pengcheng Bu and Huiwen Yan
- Subjects
RNA, Untranslated ,non-coding RNA ,Piwi-interacting RNA ,Computational biology ,Biology ,ENCODE ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Circular RNA ,Neoplasms ,microRNA ,Humans ,RNA, Small Interfering ,Molecular Biology ,Review Articles ,Diagnostics & Biomarkers ,030304 developmental biology ,Cancer ,0303 health sciences ,Gene Expression & Regulation ,long non-coding RNA ,circular RNA ,Non-coding RNA ,Long non-coding RNA ,MicroRNAs ,piwi RNA ,030220 oncology & carcinogenesis ,Cell Cycle, Growth & Proliferation ,RNA ,Human genome ,RNA, Long Noncoding ,Function (biology) - Abstract
Majority of the human genome is transcribed to RNAs that do not encode proteins. These non-coding RNAs (ncRNAs) play crucial roles in regulating the initiation and progression of various cancers. Given the importance of the ncRNAs, the roles of ncRNAs in cancers have been reviewed elsewhere. Thus, in this review, we mainly focus on the recent studies of the function, regulatory mechanism and therapeutic potential of the ncRNAs including microRNA (miRNA), long ncRNA (lncRNA), circular RNA (circRNA) and PIWI interacting RNA (piRNA), in different type of cancers.
- Published
- 2020
16. N
- Author
-
Xinyu, Wang, Chong, Liu, Siwei, Zhang, Huiwen, Yan, Liwen, Zhang, Amin, Jiang, Yong, Liu, Yun, Feng, Di, Li, Yuting, Guo, Xinyao, Hu, Yajing, Lin, Pengcheng, Bu, and Dong, Li
- Subjects
Cell Nucleus ,Adenosine ,Lung Neoplasms ,Esophageal Neoplasms ,Apoptosis ,Mice, SCID ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Mice ,Mice, Inbred NOD ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Animals ,Humans ,Female ,RNA, Long Noncoding ,Esophageal Squamous Cell Carcinoma ,Cell Proliferation - Abstract
N6-methyladenosine (m
- Published
- 2020
17. Fusobacterium nucleatum Promotes Colorectal Cancer Cell to Acquire Stem Cell‐Like Features by Manipulating Lipid Droplet‐Mediated Numb Degradation
- Author
-
Haiyang Liu, Junfeng Du, Shanshan Chao, Shuoguo Li, Huiyun Cai, Hongjie Zhang, Gang Chen, Pingsheng Liu, and Pengcheng Bu
- Subjects
Fusobacterium nucleatum ,Stem Cells ,General Chemical Engineering ,Fatty Acids ,General Engineering ,Membrane Proteins ,General Physics and Astronomy ,Medicine (miscellaneous) ,Nerve Tissue Proteins ,Lipid Droplets ,Lipids ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Fusobacterium Infections ,Humans ,General Materials Science ,Colorectal Neoplasms - Abstract
Fusobacterium nucleatum is a critical microbe that contributes to colorectal cancer progression and chemoresistance. However, whether and how F. nucleatum regulates colorectal cancer stem-like cells (CCSCs) remains unknown. Here, the authors show that F. nucleatum promotes CCSC self-renewal, and non-CCSCs to acquire CCSC features by manipulating cellular lipid accumulation. F. nucleatum infection decreases lipid accumulation in CCSCs by enhancing fatty acid oxidation, thus promoting CCSC self-renewal. In contrast, F. nucleatum increases lipid accumulation in non-CCSCs by promoting fatty acid formation. Lipids are deposited as lipid droplets, which recruits Numb, a key cell fate regulator, through the AP2A/ACSL3 complex, and MDM2, an E3 ubiquitin ligase, though VCP and UBXD8. On lipid droplets, Numb is degraded by MDM2, activating Notch signaling, thus promoting gain of stem-like cell features. Their findings demonstrate that F. nucleatum directly manipulates colorectal cancer cell fate and reveal the mechanism of lipid droplet-mediated Numb degradation for activating Notch signaling.
- Published
- 2022
18. Creatine promotes cancer metastasis through activation of Smad2/3
- Author
-
Wen Wang, Liwen Zhang, Zhenzhen Wu, David S. Hsu, Zijing Zhu, Junfeng Du, Fei Zhang, Huiyun Cai, Guizhi Shi, Xuanxuan Zhang, Pu Gao, Hai-long Piao, Qixiang Zhang, Pengcheng Bu, Huiwen Yan, and Gang Chen
- Subjects
0301 basic medicine ,Physiology ,Slug ,Colorectal cancer ,Breast Neoplasms ,Smad2 Protein ,Creatine ,Cell Line ,Metastasis ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,Downregulation and upregulation ,medicine ,Animals ,Humans ,Smad3 Protein ,Molecular Biology ,Mice, Inbred BALB C ,Gene knockdown ,biology ,business.industry ,Cell Biology ,Transforming growth factor beta ,biology.organism_classification ,medicine.disease ,030104 developmental biology ,chemistry ,Dietary Supplements ,Cancer research ,biology.protein ,Female ,Colorectal Neoplasms ,business ,030217 neurology & neurosurgery - Abstract
As one of the most popular nutrient supplements, creatine has been highly used to increase muscle mass and improve exercise performance. Here, we report an adverse effect of creatine using orthotopic mouse models, showing that creatine promotes colorectal and breast cancer metastasis and shortens mouse survival. We show that glycine amidinotransferase (GATM), the rate-limiting enzyme for creatine synthesis, is upregulated in liver metastases. Dietary uptake, or GATM-mediated de novo synthesis of creatine, enhances cancer metastasis and shortens mouse survival by upregulation of Snail and Slug expression via monopolar spindle 1 (MPS1)-activated Smad2 and Smad3 phosphorylation. GATM knockdown or MPS1 inhibition suppresses cancer metastasis and benefits mouse survival by downregulating Snail and Slug. Our findings call for using caution when considering dietary creatine to improve muscle mass or treat diseases and suggest that targeting GATM or MPS1 prevents cancer metastasis, especially metastasis of transforming growth factor beta receptor mutant colorectal cancers.
- Published
- 2021
19. miR-34a is a microRNA safeguard for Citrobacter-induced inflammatory colon oncogenesis
- Author
-
Kai-Yuan Chen, Ergang Wang, Gaiting Zhou, Yi Wang, Zhenzhen Wu, Hong Shen, Huiwen Yan, Nikolai Rakhilin, Jeffrey I. Everitt, Lihua Wang, Xiling Shen, Zhiguo Sun, Kun Xiang, Robert Mines, Gaoqi Ye, Shanshan Chao, and Pengcheng Bu
- Subjects
0301 basic medicine ,Mouse ,QH301-705.5 ,Cellular differentiation ,Science ,Inflammation ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,microRNA ,medicine ,Biology (General) ,Tissue homeostasis ,Cancer Biology ,Orphan receptor ,General Immunology and Microbiology ,General Neuroscience ,Interleukin ,General Medicine ,stem cell ,030104 developmental biology ,colon cancer ,inflammation ,Cancer research ,Medicine ,Citrobacter rodentium ,Th17 ,Stem cell ,medicine.symptom ,Carcinogenesis ,Research Article - Abstract
Inflammation often induces regeneration to repair the tissue damage. However, chronic inflammation can transform temporary hyperplasia into a fertile ground for tumorigenesis. Here, we demonstrate that the microRNA miR-34a acts as a central safeguard to protect the inflammatory stem cell niche and reparative regeneration. Although playing little role in regular homeostasis, miR-34a deficiency leads to colon tumorigenesis after Citrobacter rodentium infection. miR-34a targets both immune and epithelial cells to restrain inflammation-induced stem cell proliferation. miR-34a targets Interleukin six receptor (IL-6R) and Interleukin 23 receptor (IL-23R) to suppress T helper 17 (Th17) cell differentiation and expansion, targets chemokine CCL22 to hinder Th17 cell recruitment to the colon epithelium, and targets an orphan receptor Interleukin 17 receptor D (IL-17RD) to inhibit IL-17-induced stem cell proliferation. Our study highlights the importance of microRNAs in protecting the stem cell niche during inflammation despite their lack of function in regular tissue homeostasis.
- Published
- 2018
20. N6-methyladenosine modification of MALAT1 promotes metastasis via reshaping nuclear speckles
- Author
-
Xinyu Wang, Siwei Zhang, Chong Liu, Yuting Guo, Amin Jiang, Yajing Lin, Dong Li, Yun Feng, Huiwen Yan, Pengcheng Bu, Liwen Zhang, Xinyao Hu, Di Li, and Liu Yong
- Subjects
0303 health sciences ,MALAT1 ,Methyltransferase complex ,Mutant ,RNA ,Cell Biology ,Biology ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,Metastasis ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,chemistry ,Cancer cell ,medicine ,Binding site ,N6-Methyladenosine ,Molecular Biology ,030217 neurology & neurosurgery ,030304 developmental biology ,Developmental Biology - Abstract
Summary N6-methyladenosine (m6A), one of the most prevalent RNA post-transcriptional modifications, is involved in numerous biological processes. In previous studies, the functions of m6A were typically identified by perturbing the activity of the methyltransferase complex. Here, we dissect the contribution of m6A to an individual-long noncoding RNA—metastasis-associated lung adenocarcinoma transcript 1 (MALAT1). The mutant MALAT1 lacking m6A-motifs significantly suppressed the metastatic potential of cancer cells both in vitro and in vivo in mouse. Super-resolution imaging showed that the concatenated m6A residues on MALAT1 acted as a scaffold for recruiting YTH-domain-containing protein 1 (YTHDC1) to nuclear speckles. We further reveal that the recognition of MALAT1-m6A by YTHDC1 played a critical role in maintaining the composition and genomic binding sites of nuclear speckles, which regulate the expression of several key oncogenes. Furthermore, artificially tethering YTHDC1 onto m6A-deficient MALAT1 largely rescues the metastatic potential of cancer cells.
- Published
- 2021
21. A miR-34a-Numb Feedforward Loop Triggered by Inflammation Regulates Asymmetric Stem Cell Division in Intestine and Colon Cancer
- Author
-
Kai-Yuan Chen, Yiwei Ai, Preetish Kadur Lakshminarasimha Murthy, Pengcheng Bu, Huanhuan Joyce Chen, Anastasia Kristine Varanko, Tara Srinivasan, Sarah King, Lihua Wang, Steven M. Lipkin, Kuei-Ling Tung, and Xiling Shen
- Subjects
0301 basic medicine ,Cellular differentiation ,Molecular Sequence Data ,Population ,Nerve Tissue Proteins ,Biology ,Cell fate determination ,Article ,Mice ,03 medical and health sciences ,Stress, Physiological ,Medicine and Health Sciences ,Genetics ,Asymmetric cell division ,Animals ,Cell Lineage ,education ,Cell Proliferation ,Asymmetric stem cell division ,Inflammation ,education.field_of_study ,Base Sequence ,Receptors, Notch ,Tumor Necrosis Factor-alpha ,Cell growth ,Asymmetric Cell Division ,Membrane Proteins ,Cell Differentiation ,Cell Biology ,Cell biology ,MicroRNAs ,Editorial ,030104 developmental biology ,Gene Knockdown Techniques ,Neoplastic Stem Cells ,NUMB ,Molecular Medicine ,Stem cell - Abstract
Emerging evidence suggests that microRNAs can initiate asymmetric division, but whether microRNA and protein cell fate determinants coordinate with each other remains unclear. Here, we show that miR-34a directly suppresses Numb in early-stage colon cancer stem cells (CCSCs), forming an incoherent feedforward loop (IFFL) targeting Notch to separate stem and non-stem cell fates robustly. Perturbation of the IFFL leads to a new intermediate cell population with plastic and ambiguous identity. Lgr5+ mouse intestinal/colon stem cells (ISCs) predominantly undergo symmetric division but turn on asymmetric division to curb the number of ISCs when proinflammatory response causes excessive proliferation. Deletion of miR-34a inhibits asymmetric division and exacerbates Lgr5+ ISC proliferation under such stress. Collectively, our data indicate that microRNA and protein cell fate determinants coordinate to enhance robustness of cell fate decision, and they provide a safeguard mechanism against stem cell proliferation induced by inflammation or oncogenic mutation.
- Published
- 2016
22. Targeted drug delivery to circulating tumor cells via platelet membrane-functionalized particles
- Author
-
Xiling Shen, Jiahe Li, Charles C. Sharkey, Michael R. King, Qianhui Wu, Lihua Wang, Yiwei Ai, Pengcheng Bu, Sweta Roy, and Brittany Wun
- Subjects
Blood Platelets ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,Biophysics ,Antineoplastic Agents ,Bioengineering ,02 engineering and technology ,Biology ,Article ,Metastasis ,Biomaterials ,Mice ,03 medical and health sciences ,Drug Delivery Systems ,Circulating tumor cell ,Phagocytosis ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Platelet activation ,Tumor microenvironment ,Cell Membrane ,Cancer ,Neoplastic Cells, Circulating ,021001 nanoscience & nanotechnology ,medicine.disease ,030104 developmental biology ,Cytokine ,Targeted drug delivery ,Mechanics of Materials ,Cancer cell ,Ceramics and Composites ,Cancer research ,0210 nano-technology - Abstract
Circulating tumor cells (CTCs) are responsible for metastases in distant organs via hematogenous dissemination. Fundamental studies in the past decade have suggested that neutralization of CTCs in circulation could represent an effective strategy to prevent metastasis. Current paradigms of targeted drug delivery into a solid tumor largely fall into two main categories: unique cancer markers (e.g. overexpression of surface receptors) and tumor-specific microenvironment (e.g. low pH, hypoxia, etc.). While relying on a surface receptor to target CTCs can be greatly challenged by cancer heterogeneity, targeting of tumor microenvironments has the advantage of recognizing a broader spectrum of cancer cells regardless of genetic differences or tumor types. The blood circulation, however, where CTCs transit through, lacks the same tumor microenvironment as that found in a solid tumor. In this study, a unique “microenvironment” was confirmed upon introduction of cancer cells of different types into circulation where activated platelets and fibrin were physically associated with blood-borne cancer cells. Inspired by this observation, synthetic silica particles were functionalized with activated platelet membrane along with surface conjugation of tumor-specific apoptosis-inducing ligand cytokine, TRAIL. Biomimetic synthetic particles incorporated into CTC-associated micro-thrombi in lung vasculature and dramatically decreased lung metastases in a mouse breast cancer metastasis model. Our results demonstrate a “Trojan Horse” strategy of neutralizing CTCs to attenuate metastasis.
- Published
- 2016
23. Author response: miR-34a is a microRNA safeguard for Citrobacter-induced inflammatory colon oncogenesis
- Author
-
Kun Xiang, Zhenzhen Wu, Huiwen Yan, Pengcheng Bu, Ergang Wang, Nikolai Rakhilin, Jeffrey I. Everitt, Gaiting Zhou, Yi Wang, Robert Mines, Hong Shen, Zhiguo Sun, Xiling Shen, Lihua Wang, Gaoqi Ye, Kai-Yuan Chen, and Shanshan Chao
- Subjects
0301 basic medicine ,Citrobacter ,03 medical and health sciences ,030104 developmental biology ,biology ,microRNA ,medicine ,Cancer research ,Carcinogenesis ,medicine.disease_cause ,biology.organism_classification - Published
- 2018
24. Comprehensive models of human primary and metastatic colorectal tumors in immunodeficient and immunocompetent mice by chemokine targeting
- Author
-
Christian M. Abratte, Nozomi Nishimura, Jiahn Choi, Michael L. Shuler, Zeynep H. Gümüş, Xi Kathy Zhou, Yitian Xu, Winfried Edelmann, Harry Hou, Poornima Gadamsetty, Jian Sun, Robert J. Munroe, Pengcheng Bu, Jonlin Chen, Andrew D. Miller, Nikolai Rakhilin, Robert Edwards, Govind Nandakumar, Xiling Shen, Lihua Wang, Zhiliang Huang, Randy S. Longman, Myra Arcilla, Huanhuan Joyce Chen, Kai-Yuan Chen, Daniel J. Joe, Jeffrey W. Milsom, and Steven M. Lipkin
- Subjects
Chemokine ,Somatic cell ,Colorectal cancer ,Biomedical Engineering ,Notch signaling pathway ,CCR9 ,Bioengineering ,Biology ,medicine.disease ,Applied Microbiology and Biotechnology ,3. Good health ,Metastasis ,Immune tolerance ,Chemokine receptor ,Immunology ,Medicine and Health Sciences ,biology.protein ,medicine ,Cancer research ,Molecular Medicine ,Biotechnology - Abstract
Current orthotopic xenograft models of human colorectal cancer (CRC) require surgery and do not robustly form metastases in the liver, the most common site clinically. CCR9 traffics lymphocytes to intestine and colorectum. We engineered use of the chemokine receptor CCR9 in CRC cell lines and patient-derived cells to create primary gastrointestinal (GI) tumors in immunodeficient mice by tail-vein injection rather than surgery. The tumors metastasize inducibly and robustly to the liver. Metastases have higher DKK4 and NOTCH signaling levels and are more chemoresistant than paired subcutaneous xenografts. Using this approach, we generated 17 chemokine-targeted mouse models (CTMMs) that recapitulate the majority of common human somatic CRC mutations. We also show that primary tumors can be modeled in immunocompetent mice by microinjecting CCR9-expressing cancer cell lines into early-stage mouse blastocysts, which induces central immune tolerance. We expect that CTMMs will facilitate investigation of the biology of CRC metastasis and drug screening.
- Published
- 2015
25. IRE1α is an endogenous substrate of endoplasmic-reticulum-associated degradation
- Author
-
Sander Kersten, Takamasa Inoue, Shengyi Sun, James C. Paton, John R. Yates, Xuemei Han, Takao Iwawaki, Xiling Shen, Kenneth W. Simpson, Robert D. Guber, Haoquan Wu, Guojun Shi, Yewei Ji, Adrienne W. Paton, Deyu Fang, Haibo Sha, Hongming Ma, Qiaoming Long, Hana Kim, Pengcheng Bu, Beixue Gao, Xin Shu, Ann-Hwee Lee, Gerald E. Duhamel, Ling Qi, and Billy Tsai
- Subjects
Male ,colitis ,UPR ,Endoplasmic Reticulum ,Voeding, Metabolisme en Genomica ,Mice ,Lectins ,Endoplasmic Reticulum Chaperone BiP ,Cells, Cultured ,Heat-Shock Proteins ,Oligonucleotide Array Sequence Analysis ,Mice, Knockout ,Sel1L ,Reverse Transcriptase Polymerase Chain Reaction ,Intracellular Signaling Peptides and Proteins ,Endoplasmic Reticulum-Associated Degradation ,Metabolism and Genomics ,Cell biology ,Neoplasm Proteins ,Metabolisme en Genomica ,Protein folding ,Nutrition, Metabolism and Genomics ,Female ,Protein stabilization ,Ubiquitin-Protein Ligases ,Blotting, Western ,Mice, Transgenic ,macromolecular substances ,Endoplasmic-reticulum-associated protein degradation ,Biology ,Protein Serine-Threonine Kinases ,Article ,Voeding ,Heat shock protein ,Endoribonucleases ,Life Science ,Animals ,Humans ,intestinal epithelium ,VLAG ,Nutrition ,Base Sequence ,Endoplasmic reticulum ,Gene Expression Profiling ,HEK 293 cells ,Protein turnover ,Proteins ,Cell Biology ,ERAD ,Enterocytes ,HEK293 Cells ,ER ,Hrd1 ,inflammation ,Unfolded protein response ,Unfolded Protein Response - Abstract
Endoplasmic reticulum (ER)-associated degradation (ERAD) represents a principle quality control mechanism to clear misfolded proteins in the ER; however, its physiological significance and the nature of endogenous ERAD substrates remain largely unexplored. Here we discover that IRE1α, the sensor of the unfolded protein response (UPR), is a bona fide substrate of the Sel1L-Hrd1 ERAD complex. ERAD-mediated IRE1α degradation occurs under basal conditions in a BiP-dependent manner, requires both the intramembrane hydrophilic residues of IRE1α and the lectin protein OS9, and is attenuated by ER stress. ERAD deficiency causes IRE1α protein stabilization, accumulation and mild activation both in vitro and in vivo. Although enterocyte-specific Sel1L-knockout mice (Sel1L(ΔIEC)) are viable and seem normal, they are highly susceptible to experimental colitis and inflammation-associated dysbiosis, in an IRE1α-dependent but CHOP-independent manner. Hence, Sel1L-Hrd1 ERAD serves a distinct, essential function in restraint of IRE1α signalling in vivo by managing its protein turnover.
- Published
- 2015
26. Non-coding RNAs in cancer stem cells
- Author
-
Huiwen Yan and Pengcheng Bu
- Subjects
0301 basic medicine ,Cancer Research ,RNA, Untranslated ,Tumor initiation ,Biology ,Metastasis ,03 medical and health sciences ,Cancer stem cell ,microRNA ,medicine ,Humans ,Promoter Regions, Genetic ,Transcription factor ,Cell Proliferation ,RNA ,Non-coding RNA ,medicine.disease ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,Oncology ,Cancer research ,Disease Progression ,Neoplastic Stem Cells ,Stem cell ,Transcription Factors - Abstract
Cancer stem cells (CSCs) have been shown to play a key role in tumor initiation, progression, metastasis, and therapy resistance. Despite their potential clinical importance, the mechanism of CSC regulation is not well understood. Recent evidence suggests that different types of non-coding RNAs (ncRNA), such as microRNA (miRNA) and long non-coding RNA (LncRNA), play a role in regulating CSC growth and replication by modulating transcription factors and downstream signaling pathways activated in CSCs. Here, we review the recent major findings about how they affect stem cell quality acquisition and maintenance in CSCs, as well as metastasis and therapy resistance. Drawing connections between such discoveries could be conducive to the development of novel ncRNA-based therapeutics that can selectively target CSCs and reduce rates of cancer recurrence.
- Published
- 2017
27. A Notch positive feedback in the intestinal stem cell niche is essential for stem cell self‐renewal
- Author
-
Jiahn Choi, Anastasia Kristine Varanko, Nozomi Nishimura, Kuei-Ling Tung, Tara Srinivasan, Mavee Witherspoon, Pengcheng Bu, Lihua Wang, Nikolai Rakhilin, Preetish Kadur Lakshminarasimha Murthy, Sarah King, Julio M. Belmonte, James A. Glazier, Xiling Shen, Steven M. Lipkin, and Kai-Yuan Chen
- Subjects
0301 basic medicine ,organoid ,Niche ,Notch signaling pathway ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,Mice ,intestine stem cell ,CRISPR ,Animals ,Humans ,Cell Self Renewal ,Intestinal Mucosa ,Receptor, Notch1 ,Stem Cell Niche ,Notch signaling ,Quantitative Biology & Dynamical Systems ,Positive feedback ,Genetics ,Feedback, Physiological ,Stochastic Processes ,Binding Sites ,General Immunology and Microbiology ,gene editing ,Applied Mathematics ,Stem Cells ,Systems Biology ,positive feedback ,Articles ,Intestinal epithelium ,Stem Cell Self-Renewal ,Cell biology ,Intestines ,Organoids ,030104 developmental biology ,Computational Theory and Mathematics ,Mutation ,Notch binding ,Stem cell ,General Agricultural and Biological Sciences ,Information Systems ,Signal Transduction - Abstract
The intestinal epithelium is the fastest regenerative tissue in the body, fueled by fast‐cycling stem cells. The number and identity of these dividing and migrating stem cells are maintained by a mosaic pattern at the base of the crypt. How the underlying regulatory scheme manages this dynamic stem cell niche is not entirely clear. We stimulated intestinal organoids with Notch ligands and inhibitors and discovered that intestinal stem cells employ a positive feedback mechanism via direct Notch binding to the second intron of the Notch1 gene. Inactivation of the positive feedback by CRISPR/Cas9 mutation of the binding sequence alters the mosaic stem cell niche pattern and hinders regeneration in organoids. Dynamical system analysis and agent‐based multiscale stochastic modeling suggest that the positive feedback enhances the robustness of Notch‐mediated niche patterning. This study highlights the importance of feedback mechanisms in spatiotemporal control of the stem cell niche.
- Published
- 2017
28. Author Correction: A recellularized human colon model identifies cancer driver genes
- Author
-
Leona Cohen-Gould, Jian Sun, Rita E. Serda, Steven A. Curley, Yuri Mackeyev, Michael L. Shuler, Nancy A. Jenkins, Neal G. Copeland, Lihua Wang, Xiling Shen, Pengcheng Bu, Emina H. Huang, Zhubo Wei, Asmita Bhattacharya, David J. Savage, Shuibing Chen, Steven M. Lipkin, and Huanhuan Joyce Chen
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Published Erratum ,Biomedical Engineering ,MEDLINE ,Cancer ,Bioengineering ,medicine.disease ,Applied Microbiology and Biotechnology ,Internal medicine ,Molecular Medicine ,Medicine ,business ,Gene ,Human colon ,Biotechnology - Published
- 2019
29. Spatial perturbation with synthetic protein scaffold reveals robustness of asymmetric cell division
- Author
-
Jiahe Li, Pengcheng Bu, Kai-Yuan Chen, and Xiling Shen
- Subjects
Genetics ,Scaffold protein ,0303 health sciences ,education.field_of_study ,Cell division ,Population ,Biology ,Cell fate determination ,Article ,Cell biology ,03 medical and health sciences ,Synthetic biology ,0302 clinical medicine ,Bacterial Model ,Asymmetric cell division ,Stem cell ,education ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Asymmetric cell division is an important mechanism for creating diversity in a cellular population. Stem cells commonly perform asymmetric division to generate both a daughter stem cell for self-renewal and a more differentiated daughter cell to populate the tissue. During asymmetric cell division, protein cell fate determinants asymmetrically localize to the opposite poles of a dividing cell to cause distinct cell fate. However, it remains unclear whether cell fate determination is robust to fluctuations and noise during this spatial allocation process. To answer this question, we engineered Caulobacter, a bacterial model for asymmetric division, to express synthetic scaffolds with modular protein interaction domains. These scaffolds perturbed the spatial distribution of the PleC-DivJ- DivK phospho-signaling network without changing their endogenous expression levels. Surprisingly, enforcing symmetrical distribution of these cell fate de terminants did not result in symmetric daughter fate or any morphological defects. Further computational analysis suggested that PleC and DivJ form a robust phospho-switch that can tolerate high amount of spatial variation. This insight may shed light on the presence of similar phospho-switches in stem cell asymmetric division regulation. Overall, our study demonstrates that synthetic protein scaffolds can provide a useful tool to probe biological systems for better understanding of their operating principles.
- Published
- 2013
30. A recellularized human colon model identifies cancer driver genes
- Author
-
Steven A. Curley, Michael L. Shuler, Jian Sun, Lihua Wang, Shuibing Chen, Emina H. Huang, Asmita Bhattacharya, Rita E. Serda, Yuri Mackeyev, Zhubo Wei, Neal G. Copeland, Huanhuan Joyce Chen, Pengcheng Bu, Xiling Shen, Steven M. Lipkin, Leona Cohen-Gould, Nancy A. Jenkins, and David J. Savage
- Subjects
0301 basic medicine ,Colorectal cancer ,Carcinogenesis ,Colon ,Organogenesis ,Adenomatous Polyposis Coli Protein ,Biomedical Engineering ,Mutagenesis (molecular biology technique) ,Bioengineering ,Biology ,medicine.disease_cause ,Applied Microbiology and Biotechnology ,Article ,03 medical and health sciences ,medicine ,Medicine and Health Sciences ,Humans ,Cells, Cultured ,Cell-Free System ,Tissue Engineering ,Gene Expression Profiling ,Cancer ,medicine.disease ,Sleeping Beauty transposon system ,Neoplasm Proteins ,Gene expression profiling ,030104 developmental biology ,MSH2 ,Colonic Neoplasms ,Cancer research ,Molecular Medicine ,Colorectal Neoplasms ,Ex vivo ,Biotechnology ,Genes, Neoplasm - Abstract
Refined cancer models are needed to bridge the gaps between cell line, animal and clinical research. Here we describe the engineering of an organotypic colon cancer model by recellularization of a native human matrix that contains cell-populated mucosa and an intact muscularis mucosa layer. This ex vivo system recapitulates the pathophysiological progression from APC-mutant neoplasia to submucosal invasive tumor. We used it to perform a Sleeping Beauty transposon mutagenesis screen to identify genes that cooperate with mutant APC in driving invasive neoplasia. We identified 38 candidate invasion-driver genes, 17 of which, including TCF7L2, TWIST2, MSH2, DCC, EPHB1 and EPHB2 have been previously implicated in colorectal cancer progression. Six invasion-driver genes that have not, to our knowledge, been previously described were validated in vitro using cell proliferation, migration and invasion assays and ex vivo using recellularized human colon. These results demonstrate the utility of our organoid model for studying cancer biology.
- Published
- 2016
31. Author response: A long non-coding RNA targets microRNA miR-34a to regulate colon cancer stem cell asymmetric division
- Author
-
Tara Srinivasan, Huanhuan Joyce Chen, Xiling Shen, Steven M. Lipkin, Pengcheng Bu, Kun Xiang, Yiwei Ai, and Lihua Wang
- Subjects
Colorectal cancer ,microRNA ,Cancer research ,medicine ,Biology ,Stem cell ,Division (mathematics) ,medicine.disease ,Long non-coding RNA - Published
- 2016
32. Asymmetric division: An antitumor player?
- Author
-
Xiling Shen, Pengcheng Bu, and Lihua Wang
- Subjects
0301 basic medicine ,Cancer Research ,Colorectal cancer ,Division (mathematics) ,Biology ,Cell fate determination ,medicine.disease ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,medicine ,NUMB ,Asymmetric cell division ,Molecular Medicine ,Stem cell ,Author's View - Abstract
miR-34a-mediated asymmetric cell division reins in excessive stem cell expansion during tissue regeneration in the intestine and colon. Loss of miR-34a switches asymmetric division to symmetric division and enhances stem cell proliferation. Asymmetric division also occurs in the early stages of colon cancer stem cells. Mechanistically, miR-34a, Numb, and Notch form a feed-forward loop that specifies cell fate when stem cells divide.
- Published
- 2016
33. Notch signalling regulates asymmetric division and inter-conversion between lgr5 and bmi1 expressing intestinal stem cells
- Author
-
Kai-Yuan Chen, Leonard H. Augenlicht, Elaine Bich Than, Tara Srinivasan, Kuei Ling Tung, Steven M. Lipkin, Pengcheng Bu, and Xiling Shen
- Subjects
0301 basic medicine ,inorganic chemicals ,Mice, 129 Strain ,Cell division ,Transgene ,Cell Plasticity ,Notch signaling pathway ,Mice, Transgenic ,Biology ,digestive system ,Article ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,Mice ,Proto-Oncogene Proteins ,Medicine and Health Sciences ,Animals ,Regeneration ,Cells, Cultured ,Polycomb Repressive Complex 1 ,Multidisciplinary ,Receptors, Notch ,Regeneration (biology) ,fungi ,LGR5 ,Cell biology ,Intestines ,Mice, Inbred C57BL ,Adult Stem Cells ,030104 developmental biology ,BMI1 ,Immunology ,Cell Transdifferentiation ,Stem cell ,Signal transduction ,Cell Division ,Signal Transduction - Abstract
Rapidly cycling LGR5+ intestinal stem cells (ISCs) located at the base of crypts are the primary driver of regeneration. Additionally, BMI1 expression is correlated with a slow cycling pool of ISCs located at +4 position. While previous reports have shown interconversion between these two populations following tissue injury, we provide evidence that NOTCH signaling regulates the balance between these two populations and promotes asymmetric division as a mechanism for interconversion in the mouse intestine. In both in vitro and in vivo models, NOTCH suppression reduces the ratio of BMI1+/LGR5+ ISCs while NOTCH stimulation increases this ratio. Furthermore, NOTCH signaling can activate asymmetric division after intestinal inflammation. Overall, these data provide insights into ISC plasticity, demonstrating a direct interconversion mechanism between slow- and fast-cycling ISCs.
- Published
- 2016
34. A long non-coding RNA targets microRNA miR-34a to regulate colon cancer stem cell asymmetric division
- Author
-
Xiling Shen, Tara Srinivasan, Lihua Wang, Kun Xiang, Huanhuan Joyce Chen, Yiwei Ai, Steven M. Lipkin, and Pengcheng Bu
- Subjects
0301 basic medicine ,Cellular differentiation ,non-coding RNA ,Histone Deacetylase 1 ,DNA Methyltransferase 3A ,Epigenesis, Genetic ,Medicine and Health Sciences ,DNA (Cytosine-5-)-Methyltransferases ,Biology (General) ,Promoter Regions, Genetic ,Cancer Biology ,General Neuroscience ,Stem Cells ,General Medicine ,Non-coding RNA ,Long non-coding RNA ,3. Good health ,Cell biology ,colon cancer ,Colonic Neoplasms ,Medicine ,RNA, Long Noncoding ,Stem cell ,Cell Division ,Research Article ,Human ,cancer stem cell ,QH301-705.5 ,Science ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Cancer stem cell ,Cell Line, Tumor ,microRNA ,Prohibitins ,Humans ,Gene Silencing ,General Immunology and Microbiology ,Suicide gene ,Molecular biology ,asymmetric division ,MicroRNAs ,030104 developmental biology ,Developmental Biology and Stem Cells ,Gene Expression Regulation ,Cancer cell ,methylation - Abstract
The roles of long non-coding RNAs (lncRNAs) in regulating cancer and stem cells are being increasingly appreciated. Its diverse mechanisms provide the regulatory network with a bigger repertoire to increase complexity. Here we report a novel LncRNA, Lnc34a, that is enriched in colon cancer stem cells (CCSCs) and initiates asymmetric division by directly targeting the microRNA miR-34a to cause its spatial imbalance. Lnc34a recruits Dnmt3a via PHB2 and HDAC1 to methylate and deacetylate the miR-34a promoter simultaneously, hence epigenetically silencing miR-34a expression independent of its upstream regulator, p53. Lnc34a levels affect CCSC self-renewal and colorectal cancer (CRC) growth in xenograft models. Lnc34a is upregulated in late-stage CRCs, contributing to epigenetic miR-34a silencing and CRC proliferation. The fact that lncRNA targets microRNA highlights the regulatory complexity of non-coding RNAs (ncRNAs), which occupy the bulk of the genome. DOI: http://dx.doi.org/10.7554/eLife.14620.001, eLife digest Tumors are made of millions of cells that are not all the same. A type of cancer cell known as cancer stem cells (or CSCs for short) are often better at dividing to produce new cells and moving to new sites in the body than other types of cancer cell. Very small molecules called micro ribonucleic acids (or microRNAs for short) can influence how CSCs grow and divide by regulating the activity of specific genes. For example, a microRNA molecule called miR-34a suppresses the activity of several genes – which slows the growth of various tumors, including lung and bowel cancers. This miR-34a is often missing from some types of cells in advanced tumors. Genes encode the instructions to produce RNAs, and Wang, Bu et al. wanted to find out what stops miR-34a being produced in certain bowel cancer cells. The experiments revealed a new, very long RNA molecule – named long non-coding RNA 34 (or Lnc34a) – that binds to the gene that encodes miR-34a. Lnc34a recruits proteins that modify the gene and switch off the production of miR-34a. Furthermore, microscopy experiments revealed that when colon cancer cells divide, Lnc34a is distributed unevenly so that it blocks the production of miR-34a in one daughter cell but not the other. Lastly, Wang, Bu et al. confirmed that Lnc34a is found in higher levels in CSCs than in other cancer cells, which helps them to grow and divide more rapidly. Future experiments will try to find out what controls the production of Lnc34a and search for drugs that can block this process in cancer cells. DOI: http://dx.doi.org/10.7554/eLife.14620.002
- Published
- 2016
35. miR-34 miRNAs provide a barrier for somatic cell reprogramming
- Author
-
Sang Yong Kim, Nobuhiro Okada, Lin He, Margaux J Bennett, Greg J. Hannon, Jaclyn J. Ho, Yingchao Zhong, Arzu Öztürk, Yong Jin Choi, Caifu Chen, Geoffrey G. Hicks, Chao Po Lin, Xingyue He, and Pengcheng Bu
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Male ,Homeobox protein NANOG ,Mice, 129 Strain ,Somatic cell ,Induced Pluripotent Stem Cells ,Genes, myc ,Mice, Nude ,Mice, Transgenic ,Biology ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,SOX2 ,microRNA ,Animals ,Induced pluripotent stem cell ,Transcription factor ,Psychological repression ,Cells, Cultured ,030304 developmental biology ,Homeodomain Proteins ,Mice, Knockout ,0303 health sciences ,SOXB1 Transcription Factors ,Teratoma ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Nanog Homeobox Protein ,Cell Biology ,Cellular Reprogramming ,Coculture Techniques ,Cell biology ,Mice, Inbred C57BL ,Kinetics ,MicroRNAs ,030220 oncology & carcinogenesis ,Cancer research ,Female ,RNA Interference ,Tumor Suppressor Protein p53 ,Octamer Transcription Factor-3 ,Reprogramming - Abstract
Somatic reprogramming induced by defined transcription factors is a low efficiency process that is enhanced by p53 deficiency 1-5. To date, p21 is the only p53 target shown to contribute to p53 repression of iPSC (induced pluripotent stem cell) generation 1, 3, suggesting additional p53 targets may regulate this process. Here, we demonstrated that mir-34 microRNAs (miRNAs), particularly miR-34a, exhibit p53-dependent induction during reprogramming. mir-34a deficiency in mice significantly increased reprogramming efficiency and kinetics, with miR-34a and p21 cooperatively regulating somatic reprogramming downstream of p53. Unlike p53 deficiency, which enhances reprogramming at the expense of iPSC pluripotency, genetic ablation of mir-34a promoted iPSC generation without compromising self-renewal and differentiation. Suppression of reprogramming by miR-34a was due, at least in part, to repression of pluripotency genes, including Nanog, Sox2 and Mycn (N-Myc). This post-transcriptional gene repression by miR-34a also regulated iPSC differentiation kinetics. miR-34b and c similarly repressed reprogramming; and all three mir-34 miRNAs acted cooperatively in this process. Taken together, our findings identified mir-34 miRNAs as novel p53 targets that play an essential role in restraining somatic reprogramming.
- Published
- 2011
36. Influenza virus detection with pentabody-activated nanoparticles
- Author
-
Dongling Yang, Xinglu Huang, Bin Mu, Changsheng Dong, Jing Feng, Zhixue Cheng, Jianbing Zhang, Xiyun Yan, Pengcheng Bu, and Jie Zhuang
- Subjects
medicine.drug_class ,animal diseases ,Orthomyxoviridae ,Antibodies, Viral ,Monoclonal antibody ,Sensitivity and Specificity ,Birds ,Mice ,chemistry.chemical_compound ,Virology ,Influenza, Human ,Benzoquinones ,medicine ,Animals ,Humans ,Immunoassay ,Detection limit ,biology ,medicine.diagnostic_test ,Hydroquinone ,Immunomagnetic Separation ,Antibodies, Monoclonal ,virus diseases ,biology.organism_classification ,Molecular biology ,Hydroquinones ,Quinone ,chemistry ,Influenza A virus ,Spectrophotometry ,Colloidal gold ,Influenza in Birds ,Nanoparticles ,Magnetic nanoparticles ,Oxidation-Reduction - Abstract
A nanoparticle-based immunoassay was developed for the rapid and sensitive detection of avian influenza virus (AIV). In this method, AIV-specific pentabody (pVHH3B) was conjugated to magnetic nanoparticles (MNPs) and used to capture AIV. Gold nanoparticles (GNPs), labelled with the anti-AIV mouse monoclonal antibody 3C8, were used as a detector. In the presence of target samples, the pentabody pVHH3B enriched AIV on the MNPs. Thereafter, mAb 3C8-labelled GNPs (GNPs-mAb3C8) bound to MNPs via AIV and were separated using a magnetic field. GNPs in the complex catalyzed the oxidation of hydroquinone to quinone, and the OD value of quinone was measured. The developed assay displayed substantial signal change after incubation in an AIV sample in a concentration-dependent manner. The detection limit was 10 ng/ml, which is 10 times more sensitive than conventional double-antibody sandwich enzyme-linked immunosorbent assay (DAS-ELISA). In conclusion, by combining MNPs and a novel pentabody pVHH3B, this study provided a sensitive influenza viral detection assay that has the potential to become a rapid, sensitive and inexpensive diagnostic tool for infectious diseases.
- Published
- 2010
37. Aldolase B-Mediated Fructose Metabolism Drives Metabolic Reprogramming of Colon Cancer Liver Metastasis
- Author
-
Kun Xiang, Min Lu, Jiahe Li, Liwen Zhang, Ziyang Gao, Nikolai Rakhilin, David S. Hsu, Guo-Fang Zhang, Yan Han, Inna Astapova, Christelle Johnson, Bradley B. Barth, Xiling Shen, Scott B. Crown, Mark A. Herman, Lihua Wang, Rui Xi, Yiwei Ai, Robert Mines, Pengcheng Bu, and Kai-Yuan Chen
- Subjects
0301 basic medicine ,Physiology ,Colorectal cancer ,Fructose ,Biology ,Metastasis ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Downregulation and upregulation ,Mice, Inbred NOD ,Fructose-Bisphosphate Aldolase ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Molecular Biology ,Mice, Inbred BALB C ,GATA6 ,Aldolase B ,Liver Neoplasms ,Cell Biology ,HCT116 Cells ,medicine.disease ,Primary tumor ,3. Good health ,030104 developmental biology ,chemistry ,Cancer cell ,Cancer research ,biology.protein ,Colorectal Neoplasms - Abstract
Cancer metastasis accounts for the majority of cancer-related deaths and remains a clinical challenge. Metastatic cancer cells generally resemble cells of the primary cancer, but they may be influenced by the milieu of the organs they colonize. Here, we show that colorectal cancer cells undergo metabolic reprogramming after they metastasize and colonize the liver, a key metabolic organ. In particular, via GATA6, metastatic cells in the liver upregulate the enzyme aldolase B (ALDOB), which enhances fructose metabolism and provides fuel for major pathways of central carbon metabolism during tumor cell proliferation. Targeting ALDOB or reducing dietary fructose significantly reduces liver metastatic growth but has little effect on the primary tumor. Our findings suggest that metastatic cells can take advantage of reprogrammed metabolism in their new microenvironment, especially in a metabolically active organ such as the liver. Manipulation of involved pathways may affect the course of metastatic growth.
- Published
- 2018
38. Matrix metalloproteinase inhibitors enhance the efficacy of frontline drugs against Mycobacterium tuberculosis
- Author
-
Matthew D. Zimmerman, Yitian Xu, Lihua Wang, David G. Russell, Xiling Shen, Nikolai Rakhilin, Véronique Dartois, Evgeniya V. Nazarova, Pengcheng Bu, Firat Kaya, Lu Huang, Dah-Jiun Fu, and Kai-Yuan Chen
- Subjects
Bacterial Diseases ,0301 basic medicine ,Matrix metalloproteinase inhibitor ,Antitubercular Agents ,Glycobiology ,Drug resistance ,Pharmacology ,Biochemistry ,Efficacy ,Mice ,0302 clinical medicine ,Animal Cells ,Medicine and Health Sciences ,Medicine ,Lung ,Glucans ,lcsh:QH301-705.5 ,media_common ,biology ,Pharmaceutics ,Isoniazid ,Animal Models ,3. Good health ,Actinobacteria ,Infectious Diseases ,Experimental Organism Systems ,030220 oncology & carcinogenesis ,Granulomas ,Rifampin ,Cellular Types ,Anatomy ,Research Article ,medicine.drug ,lcsh:Immunologic diseases. Allergy ,Drug ,Tuberculosis ,Granuloma, Respiratory Tract ,Immune Cells ,media_common.quotation_subject ,Immunology ,Mouse Models ,Matrix Metalloproteinase Inhibitors ,Research and Analysis Methods ,Microbiology ,Small Molecule Libraries ,Mycobacterium tuberculosis ,03 medical and health sciences ,Model Organisms ,Polysaccharides ,In vivo ,Virology ,Genetics ,Animals ,Dextran ,Molecular Biology ,Bacteria ,business.industry ,Organisms ,Biology and Life Sciences ,Proteins ,Cell Biology ,Tropical Diseases ,bacterial infections and mycoses ,biology.organism_classification ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,lcsh:Biology (General) ,Cardiovascular Anatomy ,Blood Vessels ,Parasitology ,Drug Delivery ,lcsh:RC581-607 ,business ,Collagens - Abstract
Mycobacterium tuberculosis (Mtb) remains a grave threat to world health with emerging drug resistant strains. One prominent feature of Mtb infection is the extensive reprogramming of host tissue at the site of infection. Here we report that inhibition of matrix metalloproteinase (MMP) activity by a panel of small molecule inhibitors enhances the in vivo potency of the frontline TB drugs isoniazid (INH) and rifampicin (RIF). Inhibition of MMP activity leads to an increase in pericyte-covered blood vessel numbers and appears to stabilize the integrity of the infected lung tissue. In treated mice, we observe an increased delivery and/or retention of frontline TB drugs in the infected lungs, resulting in enhanced drug efficacy. These findings indicate that targeting Mtb-induced host tissue remodeling can increase therapeutic efficacy and could enhance the effectiveness of current drug regimens., Author summary Mycobacterium tuberculosis (Mtb) continues to be the leading cause of death from a single infectious agent worldwide, leading to 1.8 million deaths in 2015. The long treatment required (6–9 months), with all of its incumbent problems, can promote the emergence of multidrug-resistant (MDR) TB strains, so strategies to shorten the treatment duration are in dire need. Mtb’s success as a pathogen hinges on its ability to remodel the host tissue, characterized by extracellular matrix (ECM) deposition and leaky vascularization. Here we report that inhibition of matrix metalloproteinases (MMPs) significantly enhances the potency of frontline TB antibiotics. These MMP inhibitors increase the relative proportion of healthy blood vessels versus leaky dysfunctional vessels at the infection site, and enhance drug delivery and/or retention. Our study highlights the potential of targeting Mtb-induced host tissue remodeling to enhance the efficacy of current frontline antibiotics. It also suggests an alternative therapeutic strategy to repair the leaky blood vessels in TB granulomas to enhance drug delivery. Repurposing of MMP inhibitors may hold the key to shortening TB treatments and combating the emergence of MDR strains.
- Published
- 2018
39. Visualization of CD146 dimerization and its regulation in living cells
- Author
-
Jie Zhuang, Dongling Yang, Xun Shen, Xiyun Yan, Jing Feng, and Pengcheng Bu
- Subjects
medicine.drug_class ,Angiogenesis ,Immunoprecipitation ,Cell Survival ,Recombinant Fusion Proteins ,CD146 Antigen ,Monoclonal antibody ,Fluorescence ,Cell Line, Tumor ,medicine ,Fluorescence Resonance Energy Transfer ,Humans ,Molecular Biology ,Tumor microenvironment ,Photobleaching ,Chemistry ,NF-kappa B ,Antibodies, Monoclonal ,Cell Biology ,Molecular biology ,Fusion protein ,Protein Transport ,Förster resonance energy transfer ,CD146 ,Culture Media, Conditioned ,Biophysics ,FRET ,Dimerization - Abstract
Our previous study showed that the adhesion molecule CD146 as a biomarker is over-expressed on activated endothelium during angiogenesis, which was induced by tumor conditional medium and inhibited by anti-CD146 monoclonal antibody (mAb AA98). However, the CD146 molecular organization on the cells is unknown. Here, using immunoprecipitation, we found that the dimerization of CD146 occurs in both normal and tumor cells. However, the dimer/monomer ratio was higher in tumor cells than in normal cells. Moreover, we found that CD146 dimerization was up-regulated by tumor conditional medium through the NF-kappa B pathway and down-regulated by mAb AA98. To further confirm that CD146 dimerization occurs in living cells, we used fluorescence resonance energy transfer (FRET) with melanoma Mel888 cells co-expressing CFP/YFP-tagged CD146 fusion proteins. By acceptor photobleaching, we observed a strong FRET signal produced by these two fluorescence-tagged proteins. The FRET efficiency reached 20.1%. Our data provide the first evidence that CD146 dimerization occurs in living cells and is regulated within the tumor microenvironment, implying that dimerization of CD146 may be associated with malignancy.
- Published
- 2007
- Full Text
- View/download PDF
40. NOTCH Signaling Regulates Asymmetric Cell Fate of Fast- and Slow-Cycling Colon Cancer-Initiating Cells
- Author
-
Leonard H. Augenlicht, Pengcheng Bu, Tara Srinivasan, Nicole C. Panarelli, Kuei Ling Tung, Xiling Shen, Jewell Walters, Elaine Bich Than, Kai-Yuan Chen, Jeffrey W. Milsom, and Steven M. Lipkin
- Subjects
0301 basic medicine ,Cancer Research ,Telomerase ,Cellular differentiation ,Notch signaling pathway ,Apoptosis ,Mice, SCID ,Biology ,Cell fate determination ,Article ,Receptors, G-Protein-Coupled ,Immunoenzyme Techniques ,03 medical and health sciences ,Mice ,Mice, Inbred NOD ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Animals ,Humans ,AC133 Antigen ,Cell Proliferation ,Polycomb Repressive Complex 1 ,Receptors, Notch ,Cell growth ,LGR5 ,Cell Differentiation ,Xenograft Model Antitumor Assays ,Cell biology ,030104 developmental biology ,Oncology ,BMI1 ,Colonic Neoplasms ,Neoplastic Stem Cells ,Signal transduction ,Signal Transduction - Abstract
Colorectal cancer cells with stem-like properties, referred to as colon cancer–initiating cells (CCIC), have high tumorigenic potential. While CCIC can differentiate to promote cellular heterogeneity, it remains unclear whether CCIC within a tumor contain distinct subpopulations. Here, we describe the co-existence of fast- and slow-cycling CCIC, which can undergo asymmetric division to generate each other, highlighting CCIC plasticity and interconvertibility. Fast-cycling CCIC express markers, such as LGR5 and CD133, rely on MYC for their proliferation, whereas slow-cycling CCIC express markers, such as BMI1 and hTERT, are independent of MYC. NOTCH signaling promotes asymmetric cell fate, regulating the balance between these two populations. Overall, our results illuminate the basis for CCIC heterogeneity and plasticity by defining a direct interconversion mechanism between slow- and fast-cycling CCIC. Cancer Res; 76(11); 3411–21. ©2016 AACR.
- Published
- 2015
41. miR-1269 promotes metastasis and forms a positive feedback loop with TGF-β
- Author
-
Yitian Xu, David S. Hsu, Bethany P. Cummings, Sarah King, Zeynep H. Gümüş, Lihua Wang, Kuei-Ling Tung, Xiling Shen, Jian Sun, Joyce Huan Chen, Adria Closa, Victor Moreno, Nikolai Rakhilin, Kai-Yuan Chen, Pengcheng Bu, Amelia S. Zessin, James R. Shealy, Anastasia Kristine Varanko, Steven M. Lipkin, and Universitat de Barcelona
- Subjects
Male ,Colorectal cancer ,Fluorescent Antibody Technique ,General Physics and Astronomy ,Metastasis ,Mice ,Cell Movement ,Transforming Growth Factor beta ,Neoplasm Metastasis ,Càncer ,Cancer ,Aged, 80 and over ,Feedback, Physiological ,Multidisciplinary ,Middle Aged ,Prognosis ,3. Good health ,Real-time polymerase chain reaction ,Female ,Cancer chemotherapy ,Colorectal Neoplasms ,HT29 Cells ,Adult ,Chromatin Immunoprecipitation ,Blotting, Western ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,General Biochemistry, Genetics and Molecular Biology ,SOXC Transcription Factors ,Smad7 Protein ,Young Adult ,Quimioteràpia del càncer ,SOX4 ,Metàstasi ,Downregulation and upregulation ,Cell Line, Tumor ,microRNA ,medicine ,Animals ,Humans ,Aged ,Cell Proliferation ,Neoplasm Staging ,Homeodomain Proteins ,General Chemistry ,HCT116 Cells ,medicine.disease ,MicroRNAs ,Immunology ,Cancer research ,Neoplasm Recurrence, Local ,Neoplasm Transplantation ,HOXD10 ,Transcription Factors ,Transforming growth factor - Abstract
As patient survival drops precipitously from early-stage cancers to late-stage and metastatic cancers, microRNAs that promote relapse and metastasis can serve as prognostic and predictive markers as well as therapeutic targets for chemoprevention. Here we show that miR-1269a promotes colorectal cancer (CRC) metastasis and forms a positive feedback loop with TGF-β signalling. miR-1269a is upregulated in late-stage CRCs, and long-term monitoring of 100 stage II CRC patients revealed that miR-1269a expression in their surgically removed primary tumours is strongly associated with risk of CRC relapse and metastasis. Consistent with clinical observations, miR-1269a significantly increases the ability of CRC cells to invade and metastasize in vivo. TGF-β activates miR-1269 via Sox4, while miR-1269a enhances TGF-β signalling by targeting Smad7 and HOXD10, hence forming a positive feedback loop. Our findings suggest that miR-1269a is a potential marker to inform adjuvant chemotherapy decisions for CRC patients and a potential therapeutic target to deter metastasis.
- Published
- 2015
42. Understanding the Dynamics of the Stem Cell Niche in the Small Intestine with Femtosecond Laser Photodisruption
- Author
-
Jiahn Choi, Nikolai Rakhilin, Xiling Shen, Poornima Gadamsetty, Nozomi Nishimura, Daniel J. Joe, Steven M. Lipkin, and Pengcheng Bu
- Subjects
Laser ablation ,Photodisruption ,Chemistry ,Laser ,Small intestine ,Green fluorescent protein ,law.invention ,medicine.anatomical_structure ,In vivo ,law ,Femtosecond ,Biophysics ,medicine ,Stem cell - Abstract
In the stem cell niche, contact between different cells is thought to regulate stem cell self-renewal. We investigate how lost stem cells are replaced by ablating cells with in vivo femtosecond laser photodisruption in mice.
- Published
- 2015
43. Anti-CD146 monoclonal antibody AA98 inhibits angiogenesis via suppression of nuclear factor-κB activation
- Author
-
Jie Zhuang, Xiyun Yan, Jing Feng, Lizeng Gao, Pengcheng Bu, and Dongling Yang
- Subjects
Cancer Research ,Angiogenesis ,p38 mitogen-activated protein kinases ,Interleukin-1beta ,Intercellular Adhesion Molecule-1 ,Down-Regulation ,Angiogenesis Inhibitors ,Antineoplastic Agents ,CD146 Antigen ,Biology ,p38 Mitogen-Activated Protein Kinases ,Cell Line ,Cell Line, Tumor ,Humans ,Phosphorylation ,Protein kinase A ,Microscopy, Confocal ,Tumor Necrosis Factor-alpha ,NF-kappa B ,Antibodies, Monoclonal ,Endothelial Cells ,Cell migration ,Cell biology ,Endothelial stem cell ,Vascular endothelial growth factor A ,Matrix Metalloproteinase 9 ,Oncology ,CD146 ,Female - Abstract
Our previous study showed that an anti-CD146 monoclonal antibody (mAb), AA98, which was raised against the vascular endothelial cells stimulated by a conditioned medium from hepatocarcinoma SMMC 7721 cells (SMMC 7721-CM), inhibited cell migration, angiogenesis, and tumor growth. However, the underlying mechanism was not elucidated. The objective of this study was to understand the mechanism by which mAb AA98 inhibits the endothelial cell migration and angiogenesis that is induced by SMMC 7721-CM. Using confocal imaging and biochemical studies, we found that SMMC 7721-CM induced nuclear factor κB (NF-κB) activation through the upstream p38 mitogen-activated protein kinase pathway, leading to the up-regulation of matrix metalloproteinase 9 and intercellular adhesion molecule 1 expression. Interestingly, all these activities stimulated by SMMC 7721-CM could be effectively inhibited by mAb AA98 in a dose- and time-dependent manner. Our data showed that the engagement of mAb AA98 with membrane protein CD146 inhibited p38 mitogen-activated protein kinase phosphorylation, suppressed NF-κB activation, and down-regulated matrix metalloproteinase 9 and intercellular adhesion molecule 1 expression, suggesting that the suppression of NF-κB is a critical point for the inhibitory function of mAb AA98 on endothelial cell migration, angiogenesis, and tumor metastasis. These results will provide clues for a better understanding of the mechanisms underlying tumor angiogenesis as well as antiangiogenesis therapy. [Mol Cancer Ther 2006;5(11):2872–8]
- Published
- 2006
44. The expression of GFP under the control of fibroin promotor in primary ovarian cells ofAntheraea pernyi
- Author
-
Lijia An, Pengcheng Bu, Wenli Li, and Liji Jin
- Subjects
Green Fluorescent Proteins ,Molecular Sequence Data ,Fibroin ,Antheraea pernyi ,General Biochemistry, Genetics and Molecular Biology ,Green fluorescent protein ,Rapid amplification of cDNA ends ,Western blot ,Sequence Homology, Nucleic Acid ,medicine ,Animals ,Promoter Regions, Genetic ,Gene ,Expression vector ,Base Sequence ,medicine.diagnostic_test ,biology ,Ovary ,fungi ,Promoter ,DNA ,General Medicine ,Bombyx ,biology.organism_classification ,Molecular biology ,Luminescent Proteins ,Electrophoresis, Polyacrylamide Gel ,Female ,Fibroins ,General Agricultural and Biological Sciences - Abstract
The fibroin promoter can stably express foreign gene in lepidopteran cells. Total RNA was extracted from the gland of silkworm, Antheraea pernyi and the transcription initiation site of fibroin gene of A. pernyi was identified by RNA ligase mediated rapid amplification of cDNA ends (RLM-RACE). The expression vector (pGFP-N2/Fib) was constructed by use of replacing the CMV promoter with the fibroin promoter. The results of visual screening under a fluorescent inverted microscope and Western blot analysis indicated that the GFP gene was expressed in the primary cells of ovary origins from A. pernyi.
- Published
- 2003
45. A Metabolic Signature of Colon Cancer Initiating Cells
- Author
-
Nikolai Rakhilin, Pengcheng Bu, Kai-Yuan Chen, Xiaojing Liu, Xiling Shen, Chieh-Sheng Lin, and Jason W. Locasale
- Subjects
Colorectal cancer ,Down-Regulation ,Biology ,Bioinformatics ,Article ,Mass Spectrometry ,Transcriptome ,Metabolomics ,Methionine ,Downregulation and upregulation ,Gene Frequency ,Antigens, CD ,Cell Line, Tumor ,medicine ,Metabolome ,Biomarkers, Tumor ,Humans ,Glycolysis ,Epigenetics ,AC133 Antigen ,Cysteine ,neoplasms ,Chromatography, High Pressure Liquid ,Glycoproteins ,medicine.disease ,Phenotype ,Colonic Neoplasms ,Cancer research ,Neoplastic Stem Cells ,Carbohydrate Metabolism ,Peptides - Abstract
Colon cancer initiating cells (CCICs) are more tumorigenic and metastatic than the majority of colorectal cancer (CRC) cells. CCICs have also been associated with stem cell-like properties. However, there is a lack of system-level understanding of what mechanisms distinguish CCICs from common CRC cells. We compared the transcriptomes of CD133+ CCICs and CD133- CRC cells from multiple sources, which identified a distinct metabolic signature for CD133(high) CCICs. High-resolution unbiased metabolomics was then performed to validate this CCIC metabolic signature. Specifically, levels of enzymes and metabolites involved in glycolysis, the citric acid (TCA) cycle, and cysteine and methionine metabolism are altered in CCICs. Analyses of the alterations further suggest an epigenetic link. This metabolic signature provides mechanistic insights into CCIC phenotypes and may serve as potential biomarkers and therapeutic targets for future CRC treatment.
- Published
- 2014
46. A positive feedback between p53 and miR-34 miRNAs mediates tumor suppression
- Author
-
Lin He, Marcelo C. Ribeiro, Xingyue He, Terry P. Speed, Biao He, Gregory Lai, Anne Biton, J. Erby Wilkinson, Nobuhiro Okada, David M. Jablons, Hannes Vogel, Pengcheng Bu, Andreas Keller, and Chao Po Lin
- Subjects
Untranslated region ,p53 ,Lung Neoplasms ,Polyadenylation ,Haploinsufficiency ,medicine.disease_cause ,Medical and Health Sciences ,law.invention ,Mice ,law ,Tumor Cells, Cultured ,2.1 Biological and endogenous factors ,Aetiology ,Lung ,Cancer ,Feedback, Physiological ,Tumor ,Cultured ,Lung Cancer ,Biological Sciences ,Cell biology ,Tumor Cells ,microRNAs ,Gene Expression Regulation, Neoplastic ,Research Paper ,Biotechnology ,Gene isoform ,Physiological ,Context (language use) ,Adenocarcinoma of Lung ,Biology ,Adenocarcinoma ,Cell Line ,Feedback ,Mdm4 ,Cell Line, Tumor ,Proto-Oncogene Proteins ,microRNA ,medicine ,Genetics ,Animals ,Humans ,Positive feedback ,Neoplastic ,miR-34 ,Psychology and Cognitive Sciences ,Molecular biology ,MicroRNAs ,Gene Expression Regulation ,ras Proteins ,Suppressor ,Tumor Suppressor Protein p53 ,Carcinogenesis ,Gene Deletion ,HDM4 ,Developmental Biology - Abstract
As bona fide p53 transcriptional targets, miR-34 microRNAs (miRNAs) exhibit frequent alterations in many human tumor types and elicit multiple p53 downstream effects upon overexpression. Unexpectedly, miR-34 deletion alone fails to impair multiple p53-mediated tumor suppressor effects in mice, possibly due to the considerable redundancy in the p53 pathway. Here, we demonstrate that miR-34a represses HDM4, a potent negative regulator of p53, creating a positive feedback loop acting on p53. In a Kras-induced mouse lung cancer model, miR-34a deficiency alone does not exhibit a strong oncogenic effect. However, miR-34a deficiency strongly promotes tumorigenesis when p53 is haploinsufficient, suggesting that the defective p53–miR-34 feedback loop can enhance oncogenesis in a specific context. The importance of the p53/miR-34/HDM4 feedback loop is further confirmed by an inverse correlation between miR-34 and full-length HDM4 in human lung adenocarcinomas. In addition, human lung adenocarcinomas generate an elevated level of a short HDM4 isoform through alternative polyadenylation. This short HDM4 isoform lacks miR-34-binding sites in the 3′ untranslated region (UTR), thereby evading miR-34 regulation to disable the p53-miR-34 positive feedback. Taken together, our results elucidated the intricate cross-talk between p53 and miR-34 miRNAs and revealed an important tumor suppressor effect generated by this positive feedback loop.
- Published
- 2014
47. Targeting endothelial CD146 attenuates colitis and prevents colitis-associated carcinogenesis
- Author
-
Zhihua Liu, Jing Feng, Jing Yang, Hongxia Duan, Dan Liu, Dongling Yang, Zhihai Qin, Ping Wang, Pengcheng Bu, Yongting Luo, Shu Xing, Xiyun Yan, and Lina Song
- Subjects
Transcriptional Activation ,Endothelium ,Angiogenesis ,Carcinogenesis ,medicine.medical_treatment ,Interleukin-1beta ,Inflammation ,CD146 Antigen ,Cell Communication ,Inflammatory bowel disease ,Antibodies ,Pathology and Forensic Medicine ,Proinflammatory cytokine ,medicine ,Human Umbilical Vein Endothelial Cells ,Leukocytes ,Animals ,Humans ,Colitis ,Mice, Knockout ,Neovascularization, Pathologic ,business.industry ,Tumor Necrosis Factor-alpha ,Dextran Sulfate ,NF-kappa B ,Endothelial Cells ,medicine.disease ,Up-Regulation ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,Cytokine ,Immunology ,Tumor necrosis factor alpha ,medicine.symptom ,business ,Colorectal Neoplasms - Abstract
Recently, enhanced CD146 expression was reported on endothelial cells in intestinal biopsies from patients with inflammatory bowel disease. However, the underlying mechanism remains unknown. Here, we found that overexpressed endothelial CD146 promoted the inflammatory responses in inflammatory bowel disease, which further potentiated the occurrence of colitis-associated colorectal carcinogenesis. Eliminating endothelial CD146 by conditional knockout significantly ameliorated the severity of inflammation in two different murine models of colitis, and decreased tumor incidence and tumor progression in a murine model of colitis-associated colorectal carcinogenesis. Mechanistic study showed that cytokine tumor necrosis factor-α (TNF-α) up-regulated the expression of endothelial CD146 through NF-κB transactivation. In turn, the enhanced endothelial CD146 expression promoted both angiogenesis and proinflammatory leukocyte extravasations, contributing to inflammation. Using an anti-CD146 antibody, AA98, alone or together with an anti–TNF-α antibody significantly attenuated colitis and prevented colitis-associated colorectal carcinogenesis in mice. Our study provides the first evidence that CD146 plays a dual role on endothelium, facilitating leukocyte extravasations and angiogenesis, thus promoting inflammation. This finding not only reveals the function and regulating mechanism of CD146 in inflammatory bowel disease, but also provides a promising therapeutic strategy for treating inflammatory bowel disease and preventing colitis-associated colorectal carcinogenesis.
- Published
- 2013
48. Asymmetric division: a marker for cancer stem cells in early stage tumors?
- Author
-
Pengcheng, Bu, Kai-Yuan, Chen, Steven M, Lipkin, and Xiling, Shen
- Subjects
Editorial ,Neoplasms ,Asymmetric Cell Division ,Neoplastic Stem Cells ,Humans ,Neoplasm Staging - Published
- 2013
49. Chemokine 25–induced signaling suppresses colon cancer invasion and metastasis
- Author
-
Robert Edwards, Serena Tucci, Xiling Shen, Huanhuan Joyce Chen, Jeffrey W. Milsom, Pengcheng Bu, Sang W. Lee, Steven M. Lipkin, Zeynep H. Gümüş, and Winfried Edelmann
- Subjects
Chemokine ,Colorectal cancer ,Notch signaling pathway ,CCR9 ,Mice, SCID ,Adenocarcinoma ,Metastasis ,Chemokine receptor ,Mice ,Receptors, CCR ,Mice, Inbred NOD ,Cell Line, Tumor ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Gene Regulatory Networks ,Neoplasm Invasiveness ,Homeodomain Proteins ,biology ,Chemotaxis ,General Medicine ,medicine.disease ,Primary tumor ,digestive system diseases ,Gene Expression Regulation, Neoplastic ,Chemokines, CC ,Cancer research ,biology.protein ,Transcription Factor HES-1 ,CCL25 ,Colorectal Neoplasms ,Neoplasm Transplantation ,Research Article ,Signal Transduction - Abstract
Chemotactic cytokines (chemokines) can help regulate tumor cell invasion and metastasis. Here, we show that chemokine 25 (CCL25) and its cognate receptor chemokine receptor 9 (CCR9) inhibit colorectal cancer (CRC) invasion and metastasis. We found that CCR9 protein expression levels were highest in colon adenomas and progressively decreased in invasive and metastatic CRCs. CCR9 was expressed in both primary tumor cell cultures and colon-cancer-initiating cell (CCIC) lines derived from early-stage CRCs but not from metastatic CRC. CCL25 stimulated cell proliferation by activating AKT signaling. In vivo, systemically injected CCR9+ early-stage CCICs led to the formation of orthotopic gastrointestinal xenograft tumors. Blocking CCR9 signaling inhibited CRC tumor formation in the native gastrointestinal CCL25+ microenvironment, while increasing extraintestinal tumor incidence. NOTCH signaling, which promotes CRC metastasis, increased extraintestinal tumor frequency by stimulating CCR9 proteasomal degradation. Overall, these data indicate that CCL25 and CCR9 regulate CRC progression and invasion and further demonstrate an appropriate in vivo experimental system to study CRC progression in the native colon microenvironment.
- Published
- 2012
50. A novel antibody AA98 V(H)/L directed against CD146 efficiently inhibits angiogenesis
- Author
-
Yun, Lin, Xiaoping, Wu, Yi, Shen, Pengcheng, Bu, Dongling, Yang, and Xiyun, Yan
- Subjects
Epitopes ,Immunoglobulin Variable Region ,Animals ,Antibodies, Monoclonal ,Endothelial Cells ,Humans ,Neovascularization, Physiologic ,Immunoglobulin Light Chains ,CD146 Antigen ,Chick Embryo ,Immunoglobulin Heavy Chains ,Chorioallantoic Membrane - Abstract
An anti-CD146 monoclonal antibody, AA98, has been identified as an inhibitor of tumor angiogenesis. To overcome the inherent immunogenicity of murine antibody as well as to facilitate immunotoxin construction, a single chain AA98 V(H)/L with three-domain fragments was constructed and expressed in mammalian cells.The genes of the AA98 heavy chain variable region and the light chain were linked with a modified 12 amino acid sequence that was derived from the heavy chain C(H)1 region, thus constituting the three-domain antibody V(H)/L. Soluble AA98 V(H)/L was produced by mammalian cells and purified by affinity chromatography. The specificity of AA98 V(H)/L for the CD146 molecule was detected by ELISA, immunofluorescence staining and flow cytometry.AA98 V(H)/L alone showed anti-angiogenic properties in a chicken chorioallantoic membrane (CAM) assay as the parent mAb AA98 did.This newly generated AA98 V(H)/L antibody displays a therapeutic potential for tumor and other angiogenesis disorders, as well as providing a new strategy for antibody engineering for clinical applications.
- Published
- 2008
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.