1
|
Klagkou E, Valakos D, Foutadakis S, Polyzos A, Papadopoulou A, Vatsellas G, Thanos D. An Unbiased Approach to Identifying Cellular Reprogramming-Inducible Enhancers. Int J Mol Sci 2024; 25:13128. [PMID: 39684837 DOI: 10.3390/ijms252313128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Cellular reprogramming of somatic cells towards induced pluripotency is a multistep stochastic process mediated by the transcription factors Oct4, Sox2, Klf4 and c-Myc (OSKM), which orchestrate global epigenetic and transcriptional changes. We performed a large-scale analysis of integrated ChIP-seq, ATAC-seq and RNA-seq data and revealed the spatiotemporal highly dynamic pattern of OSKM DNA binding during reprogramming. We found that OSKM show distinct temporal patterns of binding to different classes of pluripotency-related enhancers. Genes involved in reprogramming are regulated by the coordinated activity of multiple enhancers, which are sequentially bound by OSKM for strict transcriptional control. Based on these findings, we developed an unbiased approach to identify Reprogramming-Inducible Enhancers (RIEs), constructed enhancer-traps and isolated cells undergoing reprogramming in real time. We used a representative RIE taken from the Upp1 gene fused to Gfp and isolated cells at different time-points during reprogramming and found that they have unique developmental capacities as they are reprogrammed with high efficiency due to their distinct molecular signatures. In conclusion, our experiments have led to the development of an unbiased method to identify and isolate reprogrammable cells in real time by exploiting the functional dynamics of OSKM, which can be used as efficient reprogramming biomarkers.
Collapse
Affiliation(s)
- Eleftheria Klagkou
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
- Section of Biochemistry and Molecular Biology, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, Zografou, 15772 Athens, Greece
| | - Dimitrios Valakos
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
- Section of Biochemistry and Molecular Biology, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, Zografou, 15772 Athens, Greece
| | - Spyros Foutadakis
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
- Hellenic Institute for the Study of Sepsis (HISS), 11528 Athens, Greece
| | - Alexander Polyzos
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill, Cornell Medicine, New York, NY 10065, USA
| | - Angeliki Papadopoulou
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
- Department of Computational Biology, University of Lausanne, 1015 Lausanne, Switzerland
- Department of Biology, School of Sciences and Engineering, University of Crete, 70013 Irakleio, Greece
| | - Giannis Vatsellas
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
| | - Dimitris Thanos
- Biomedical Research Foundation, Academy of Athens (BRFAA), 4 Soranou Efesiou St., 11527 Athens, Greece
| |
Collapse
|
2
|
Tsubosaka A, Komura D, Kakiuchi M, Katoh H, Onoyama T, Yamamoto A, Abe H, Seto Y, Ushiku T, Ishikawa S. Stomach encyclopedia: Combined single-cell and spatial transcriptomics reveal cell diversity and homeostatic regulation of human stomach. Cell Rep 2023; 42:113236. [PMID: 37819756 DOI: 10.1016/j.celrep.2023.113236] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/05/2023] [Accepted: 09/24/2023] [Indexed: 10/13/2023] Open
Abstract
The stomach is an important digestive organ with various biological functions. However, because of the complexity of its cellular and glandular composition, its precise cellular biology has yet to be elucidated. In this study, we conducted single-cell RNA sequencing (scRNA-seq) and subcellular-level spatial transcriptomics analysis of the human stomach and constructed the largest dataset to date: a stomach encyclopedia. This dataset consists of approximately 380,000 cells from scRNA-seq and the spatial transcriptome, enabling integrated analyses of transcriptional and spatial information of gastric and metaplastic cells. This analysis identified LEFTY1 as an uncharacterized stem cell marker, which was confirmed through lineage tracing analysis. A wide variety of cell-cell interactions between epithelial and stromal cells, including PDGFRA+BMP4+WNT5A+ fibroblasts, was highlighted in the developmental switch of intestinal metaplasia. Our extensive dataset will function as a fundamental resource in investigations of the stomach, including studies of development, aging, and carcinogenesis.
Collapse
Affiliation(s)
- Ayumu Tsubosaka
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku 1130033, Tokyo, Japan
| | - Daisuke Komura
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku 1130033, Tokyo, Japan
| | - Miwako Kakiuchi
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku 1130033, Tokyo, Japan
| | - Hiroto Katoh
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku 1130033, Tokyo, Japan
| | - Takumi Onoyama
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku 1130033, Tokyo, Japan; Division of Gastroenterology and Nephrology, Department of Multidisciplinary Internal Medicine, School of Medicine, Faculty of Medicine, Tottori University, 36-1, Nishicho, Yonago 683-8504, Tottori, Japan
| | - Asami Yamamoto
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku 1130033, Tokyo, Japan
| | - Hiroyuki Abe
- Dpartment of Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku 1130033, Tokyo, Japan
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-kyu 1130033, Tokyo, Japan
| | - Tetsuo Ushiku
- Dpartment of Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku 1130033, Tokyo, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku 1130033, Tokyo, Japan; Division of Pathology, National Cancer Center Exploratory Oncology Research & Clinical Trial Center, 6-5-1, Kashiwanoha, Kashiwa 277-8577, Chiba, Japan.
| |
Collapse
|
3
|
Monsen VT, Attramadal H. Structural insights into regulation of CCN protein activities and functions. J Cell Commun Signal 2023:10.1007/s12079-023-00768-5. [PMID: 37245184 DOI: 10.1007/s12079-023-00768-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/07/2023] [Indexed: 05/29/2023] Open
Abstract
CCN proteins play important functions during development, in repair mechanisms following tissue injury, as well as in pathophysiologic mechanisms of metastasis of cancer. CCNs are secreted proteins that have a multimodular structure and are categorized as matricellular proteins. Although the prevailing view is that CCN proteins regulate biologic processes by interacting with a wide array of other proteins in the microenvironment of the extracellular matrix, the molecular mechanisms of action of CCN proteins are still poorly understood. Not dissuading the current view, however, the recent appreciation that these proteins are signaling proteins in their own right and may even be considered preproproteins controlled by endopeptidases to release a C-terminal bioactive peptide has opened new avenues of research. Also, the recent resolution of the crystal structure of two of the domains of CCN3 have provided new knowledge with implications for the entire CCN family. These resolved structures in combination with structural predictions based upon the AlphaFold artificial intelligence tool provide means to shed new light on CCN functions in context of the notable literature in the field. CCN proteins have emerged as important therapeutic targets in several disease conditions, and clinical trials are currently ongoing. Thus, a review that critically discusses structure - function relationship of CCN proteins, in particular as it relates to interactions with other proteins in the extracellular milieu and on the cell surface, as well as to cell signaling activities of these proteins, is very timely. Suggested mechanism for activation and inhibition of signaling by the CCN protein family (graphics generated with BioRender.com ).
Collapse
Affiliation(s)
- Vivi Talstad Monsen
- Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Håvard Attramadal
- Institute for Surgical Research, Oslo University Hospital, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
4
|
Gropp M, Waldhorn I, Gil Y, Steiner D, Turetsky TT, Smith Y, Sabag O, Falick-Michaeli T, Ram SE, Reubinoff BE. Laminin111-based defined culture promoting self-renewing human pluripotent stem cells with properties of the early post-implantation epiblast. Stem Cell Reports 2022; 17:2643-2660. [PMID: 36368331 PMCID: PMC9768580 DOI: 10.1016/j.stemcr.2022.10.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/11/2022] Open
Abstract
In the mammalian embryo, a formative pluripotent phase is proposed to exist at the early post-implantation period, during the transition from the pre-implantation naive-to the post-implantation primed-epiblast. By recapitulating a laminin component of the extracellular matrix niche during embryonic formative transition, and defined culture conditions, we generated cultures highly enriched for self-renewing human pluripotent stem cells (hPSCs), exhibiting properties of early post-implantation epiblast cells. These hPSCs display post-implantation-epiblast gene expression profiles. FGF and TGF-β signaling maintain their self-renewal for multiple passages. They have inactive canonical Wnt signaling, do not express primitive streak markers, and are competent to initiate differentiation toward germline and somatic fates. hPSCs exhibiting early post-implantation epiblast properties may shed light on human embryonic PSCs development and may serve for initiating somatic and germ cell specification.
Collapse
Affiliation(s)
- Michal Gropp
- The Hadassah Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, 91120 Jerusalem, Israel
| | - Ithai Waldhorn
- The Hadassah Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, 91120 Jerusalem, Israel
| | - Yaniv Gil
- The Hadassah Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, 91120 Jerusalem, Israel
| | - Debora Steiner
- The Hadassah Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, 91120 Jerusalem, Israel
| | - Tikva Tako Turetsky
- The Hadassah Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, 91120 Jerusalem, Israel
| | - Yoav Smith
- The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ofra Sabag
- Department of Developmental Biology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Tal Falick-Michaeli
- Rubin Chair in Medical Science, Department of Developmental Biology & Cancer Research, IMRIC, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sharona Even Ram
- The Hadassah Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, 91120 Jerusalem, Israel
| | - Benjamin E. Reubinoff
- The Hadassah Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, 91120 Jerusalem, Israel,Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, 91120 Jerusalem, Israel,Corresponding author
| |
Collapse
|
5
|
Sun X, Zhang X, Yang L, Dong B. A microRNA Cluster-Lefty Pathway is Required for Cellulose Synthesis During Ascidian Larval Metamorphosis. Front Cell Dev Biol 2022; 10:835906. [PMID: 35372357 PMCID: PMC8965075 DOI: 10.3389/fcell.2022.835906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Synthesis of cellulose and formation of tunic structure are unique traits in the tunicate animal group. However, the regulatory mechanism of tunic formation remains obscure. Here, we identified a novel microRNA cluster of three microRNAs, including miR4018a, miR4000f, and miR4018b in Ciona savignyi. In situ hybridization and promoter assays showed that miR4018a/4000f/4018b cluster was expressed in the mesenchymal cells in the larval trunk, and the expression levels were downregulated during the later tailbud stage and larval metamorphosis. Importantly, overexpression of miR4018a/4000f/4018b cluster in mesenchymal cells abolished the cellulose synthesis in Ciona larvae and caused the loss of tunic cells in metamorphic larvae, indicating the regulatory roles of miR4018a/4000f/4018b cluster in cellulose synthesis and mesenchymal cell differentiation into tunic cells. To elucidate the molecular mechanism, we further identified the target genes of miR4018a/4000f/4018b cluster using the combination approaches of TargetScan prediction and RNA-seq data. Left-right determination factor (Lefty) was confirmed as one of the target genes after narrow-down screening and an experimental luciferase assay. Furthermore, we showed that Lefty was expressed in the mesenchymal and tunic cells, indicating its potentially regulatory roles in mesenchymal cell differentiation and tunic formation. Notably, the defects in tunic formation and loss of tunic cells caused by overexpression of miR4018a/4000f/4018b cluster could be restored when Lefty was overexpressed in Ciona larvae, suggesting that miR4018a/4000f/4018b regulated the differentiation of mesenchymal cells into tunic cells through the Lefty signaling pathway during ascidian metamorphosis. Our findings, thus, reveal a novel microRNA-Lefty molecular pathway that regulates mesenchymal cells differentiating into tunic cells required for the tunic formation in tunicate species.
Collapse
Affiliation(s)
- Xueping Sun
- Sars Fang Centre, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Xiaoming Zhang
- Sars Fang Centre, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Likun Yang
- Sars Fang Centre, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Bo Dong
- Sars Fang Centre, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, China
| |
Collapse
|
6
|
Changes in chromatin accessibility landscape and histone H3 core acetylation during valproic acid-induced differentiation of embryonic stem cells. Epigenetics Chromatin 2021; 14:58. [PMID: 34955095 PMCID: PMC8711205 DOI: 10.1186/s13072-021-00432-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/17/2021] [Indexed: 11/27/2022] Open
Abstract
Directed differentiation of mouse embryonic stem cells (mESCs) or induced pluripotent stem cells (iPSCs) provides powerful models to dissect the molecular mechanisms leading to the formation of specific cell lineages. Treatment with histone deacetylase inhibitors can significantly enhance the efficiency of directed differentiation. However, the mechanisms are not well understood. Here, we use CUT&RUN in combination with ATAC-seq to determine changes in both histone modifications and genome-wide chromatin accessibility following valproic acid (VPA) exposure. VPA induced a significant increase in global histone H3 acetylation (H3K56ac), a core histone modification affecting nucleosome stability, as well as enrichment at loci associated with cytoskeletal organization and cellular morphogenesis. In addition, VPA altered the levels of linker histone H1 subtypes and the total histone H1/nucleosome ratio indicative of initial differentiation events. Notably, ATAC-seq analysis revealed changes in chromatin accessibility of genes involved in regulation of CDK serine/threonine kinase activity and DNA duplex unwinding. Importantly, changes in chromatin accessibility were evident at several key genomic loci, such as the pluripotency factor Lefty, cardiac muscle troponin Tnnt2, and the homeodomain factor Hopx, which play critical roles in cardiomyocyte differentiation. Massive parallel transcription factor (TF) footprinting also indicates an increased occupancy of TFs involved in differentiation toward mesoderm and endoderm lineages and a loss of footprints of POU5F1/SOX2 pluripotency factors following VPA treatment. Our results provide the first genome-wide analysis of the chromatin landscape following VPA-induced differentiation in mESCs and provide new mechanistic insight into the intricate molecular processes that govern departure from pluripotency and early lineage commitment.
Collapse
|
7
|
Song MH, Choi SC, Noh JM, Joo HJ, Park CY, Cha JJ, Ahn TH, Ko TH, Choi JI, Na JE, Rhyu IJ, Jang Y, Park Y, Gim JA, Kim JH, Lim DS. LEFTY-PITX2 signaling pathway is critical for generation of mature and ventricular cardiac organoids in human pluripotent stem cell-derived cardiac mesoderm cells. Biomaterials 2021; 278:121133. [PMID: 34571434 DOI: 10.1016/j.biomaterials.2021.121133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023]
Abstract
The generation of mature ventricular cardiomyocytes (CMs) resembling adult CMs from human pluripotent stem cells (hPSCs) is necessary for disease modeling and drug discovery. To investigate the effect of self-organizing capacity on the generation of mature cardiac organoids (COs), we generated cardiac mesoderm cell-derived COs (CMC-COs) and CM-derived COs (CM-COs) and evaluated COs. CMC-COs exhibited more organized sarcomere structures and mitochondria, well-arranged t-tubule structures, and evenly distributed intercalated discs. Increased expressions of ventricular CM, cardiac metabolic, t-tubule formation, K+ ion channel, and junctional markers were confirmed in CMC-COs. Mature ventricular-like function such as faster motion vector speed, decreased beats per min, increased peak-to-peak duration, and prolonged APD50 and APD90 were observed in CMC-COs. Transcriptional profiling revealed that extracellular matrix-integrin, focal adhesion, and LEFTY-PITX2 signaling pathways are upregulated in CMC-COs. LEFTY knockdown affected ECM-integrin-FA signaling pathways in CMC-COs. Here, we found that high self-organizing capacity of CMCs is critical for the generation of mature and ventricular COs. We also demonstrated that LEFTY-PITX2 signaling plays key roles for CM maturation and specification into ventricular-like CM subtype in CMC-COs. CMC-COs are an attractive resource for disease modeling and drug discovery.
Collapse
Affiliation(s)
- Myeong-Hwa Song
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Seung-Cheol Choi
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea; R&D Center for Companion Diagnostic, SOL Bio Corporation, Suite 510, 27, Seongsui-ro7-gil, Seongdong-gu, Seoul, 04780, South Korea
| | - Ji-Min Noh
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Hyung Joon Joo
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Chi-Yeon Park
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Jung-Joon Cha
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Tae Hoon Ahn
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Tae Hee Ko
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, 02841, South Korea
| | - Jong-Il Choi
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, 02841, South Korea
| | - Ji Eun Na
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Im Joo Rhyu
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Yongjun Jang
- Department of Biomedical Sciences, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Yongdoo Park
- Department of Biomedical Sciences, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Jeong-An Gim
- Medical Science Research Center, College of Medicine, Korea University Guro Hospital, Seoul,08308, South Korea
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Do-Sun Lim
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea.
| |
Collapse
|
8
|
Minoshima F, Ozaki H, Odaka H, Tateno H. Integrated analysis of glycan and RNA in single cells. iScience 2021; 24:102882. [PMID: 34401666 PMCID: PMC8349903 DOI: 10.1016/j.isci.2021.102882] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/13/2021] [Accepted: 07/15/2021] [Indexed: 12/03/2022] Open
Abstract
Single-cell sequencing has emerged as an indispensable technology to dissect cellular heterogeneity but never been applied to the simultaneous analysis of glycan and RNA. Using oligonucleotide-labeled lectins, we first established lectin-based glycan profiling of single cells by sequencing (scGlycan-seq). We then combined the scGlycan-seq with single-cell transcriptome profiling for joint analysis of glycan and RNA in single cells (scGR-seq). Using scGR-seq, we analyzed the two modalities in human induced pluripotent stem cells (hiPSCs) before and after differentiation into neural progenitor cells at the single-cell resolution. The combination of RNA and glycan separated the two cell types clearer than either one of them. Furthermore, integrative analysis of glycan and RNA modalities in single cells found known and unknown lectins that were specific to hiPSCs and coordinated with neural differentiation. Taken together, we demonstrate that scGR-seq can reveal the cellular heterogeneity and biological roles of glycans across multicellular systems.
Collapse
Affiliation(s)
- Fumi Minoshima
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Haruka Ozaki
- Bioinformatics Laboratory, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Center for Artificial Intelligence Research, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Haruki Odaka
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Hiroaki Tateno
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
- JST PRESTO, Tsukuba Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| |
Collapse
|
9
|
Matsumoto T, Chino H, Akiya M, Hashimura M, Yokoi A, Tochimoto M, Nakagawa M, Jiang Z, Saegusa M. Requirements of LEFTY and Nodal overexpression for tumor cell survival under hypoxia in glioblastoma. Mol Carcinog 2020; 59:1409-1419. [PMID: 33111989 DOI: 10.1002/mc.23265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 01/06/2023]
Abstract
Glioblastomas (GBM) contain numerous hypoxic foci associated with a rare fraction of glioma stem cells (GSCs). Left-right determination factor (LEFTY) and Nodal, members of the transforming growth factor β (TGF-β) superfamily, have glycogen synthase kinase 3β (GSK-3β) phosphorylation motifs and are linked with stemness in human malignancies. Herein, we investigated the roles of LEFTY and Nodal in GBM hypoxic foci. In clinical samples, significantly higher expression of LEFTY, Nodal, phospho (p) GSK-3β, pSmad2, and Nestin, as well as higher apoptotic and lower proliferation rates, were observed in nonpseudopalisading (non-Ps) perinecrotic lesions as compared to Ps and non-necrotic tumor lesions, with a positive correlation between LEFTY, Nodal, pGSK-3β, or pSmad2 scores. In KS-1, a GBM cell line that lacks endogenous Nodal expression, treatment with the hypoxic mimetic CoCl2 increased LEFTY, pGSK-3β, and pSmad2 levels, but decreased pAkt levels. Moreover, the promoter for LEFTY, but not Nodal, was activated by Smad2 or TGF-β1, suggesting that overexpression of LEFTY and Nodal may be due to Akt-independent GSK-3β inactivation, with or without cooperation of the TGF-β1/Smad2 axis. LEFTY and Nodal overexpression increased proliferation rates and reduced susceptibility to CoCl2 -induced apoptosis, and increased the expression of epithelial-mesenchymal transition (EMT)/GSC-related markers. An increased ALDH1high population and more efficient spheroid formation was also observed in LEFTY-overexpressing cells. These findings suggest that LEFTY and Nodal may contribute to cell survival in non-Ps GBM perinecrotic lesions, leading to alterations in apoptosis, proliferation, or EMT/GCS features.
Collapse
Affiliation(s)
- Toshihide Matsumoto
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Hiromi Chino
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Masashi Akiya
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Miki Hashimura
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Ako Yokoi
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Masataka Tochimoto
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Mayu Nakagawa
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Zesong Jiang
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Makoto Saegusa
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| |
Collapse
|
10
|
Bozhokin MS, Sopova YV, Kachkin DV, Rubel AA, Khotin MG. Mechanisms of TGFβ3 Action as a Therapeutic Agent for Promoting the Synthesis of Extracellular Matrix Proteins in Hyaline Cartilage. BIOCHEMISTRY (MOSCOW) 2020; 85:436-447. [PMID: 32569551 DOI: 10.1134/s0006297920040045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Hyaline cartilage is a nonvascular connective tissue covering the joint surface. It consists mostly of the extracellular matrix proteins and a small number of highly differentiated chondrocytes. At present, various techniques for repairing joint surfaces damage, for example, the use of modified cell cultures and biodegradable scaffolds, are under investigation. Molecular mechanisms of cartilage tissue proliferation have been also actively studied in recent years. TGFβ3, which plays a critical role in the proliferation of normal cartilage tissue, is one of the most important protein among cytokines and growth factors affecting chondrogenesis. By interacting directly with receptors on the cell membrane surface, TGFβ3 triggers a cascade of molecular interactions involving transcription factor Sox9. In this review, we describe the effects of TGFβ3 on the receptor complex activation and subsequent intracellular trafficking of Smad proteins and analyze the relation between these processes and upregulation of expression of major extracellular matrix genes, such as col2a1 and acan.
Collapse
Affiliation(s)
- M S Bozhokin
- Vreden Russian Scientific Research Institute of Traumatology and Orthopedics, St. Petersburg, 195427, Russia. .,Institute of Cytology, Russian Academy of Science, St. Petersburg, 194064, Russia
| | - Y V Sopova
- Vavilov Institute of General Genetics, Russian Academy of Science, St. Petersburg Branch, St. Petersburg, 199034, Russia.,St. Petersburg State University, Faculty of Biology, St. Petersburg, 199034, Russia.,St. Petersburg State University, Laboratory of Amyloid Biology, St. Petersburg, 199034, Russia
| | - D V Kachkin
- St. Petersburg State University, Faculty of Biology, St. Petersburg, 199034, Russia.,St. Petersburg State University, Laboratory of Amyloid Biology, St. Petersburg, 199034, Russia
| | - A A Rubel
- St. Petersburg State University, Faculty of Biology, St. Petersburg, 199034, Russia.,St. Petersburg State University, Laboratory of Amyloid Biology, St. Petersburg, 199034, Russia
| | - M G Khotin
- Institute of Cytology, Russian Academy of Science, St. Petersburg, 194064, Russia
| |
Collapse
|
11
|
Griffiths R, Woods S, Cheng A, Wang P, Griffiths-Jones S, Ronshaugen M, Kimber SJ. The Transcription Factor-microRNA Regulatory Network during hESC-chondrogenesis. Sci Rep 2020; 10:4744. [PMID: 32179818 PMCID: PMC7075910 DOI: 10.1038/s41598-020-61734-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 02/19/2020] [Indexed: 12/21/2022] Open
Abstract
Human embryonic stem cells (ESCs) offer a promising therapeutic approach for osteoarthritis (OA). The unlimited source of cells capable of differentiating to chondrocytes has potential for repairing damaged cartilage or to generate disease models via gene editing. However their use is limited by the efficiency of chondrogenic differentiation. An improved understanding of the transcriptional and post-transcriptional regulation of chondrogenesis will enable us to improve hESC chondrogenic differentiation protocols. Small RNA-seq and whole transcriptome sequencing was performed on distinct stages of hESC-directed chondrogenesis. This revealed significant changes in the expression of several microRNAs including upregulation of known cartilage associated microRNAs and those transcribed from the Hox complexes, and the downregulation of pluripotency associated microRNAs. Integration of miRomes and transcriptomes generated during hESC-directed chondrogenesis identified key functionally related clusters of co-expressed microRNAs and protein coding genes, associated with pluripotency, primitive streak, limb development and extracellular matrix. Analysis identified regulators of hESC-directed chondrogenesis such as miR-29c-3p with 10 of its established targets identified as co-regulated 'ECM organisation' genes and miR-22-3p which is highly co-expressed with ECM genes and may regulate these genes indirectly by targeting the chondrogenic regulators SP1 and HDAC4. We identified several upregulated transcription factors including HOXA9/A10/D13 involved in limb patterning and RELA, JUN and NFAT5, which have targets enriched with ECM associated genes. We have developed an unbiased approach for integrating transcriptome and miRome using protein-protein interactions, transcription factor regulation and miRNA target interactions and identified key regulatory networks prominent in hESC chondrogenesis.
Collapse
Affiliation(s)
- Rosie Griffiths
- Divisions of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, Michael Smith Building, Oxford Road, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, United Kingdom
| | - Steven Woods
- Divisions of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, Michael Smith Building, Oxford Road, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Aixin Cheng
- Divisions of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, Michael Smith Building, Oxford Road, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
- Salford Royal NHS Foundation Trust, Department of Trauma and Orthopaedic, Stott Lane, Salford, M6 8HD, United Kingdom
| | - Ping Wang
- Evolution and Genomic Science, Faculty of Biology Medicine and Health, Michael Smith Building, Oxford Road, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Sam Griffiths-Jones
- Evolution and Genomic Science, Faculty of Biology Medicine and Health, Michael Smith Building, Oxford Road, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Matthew Ronshaugen
- Developmental Biology and Medicine, Faculty of Biology Medicine and Health, Michael Smith Building, Oxford Road, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Susan J Kimber
- Divisions of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, Michael Smith Building, Oxford Road, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK.
| |
Collapse
|
12
|
Zhang L, Liu X, Liang J, Wu J, Tan D, Hu W. Lefty-1 inhibits renal epithelial-mesenchymal transition by antagonizing the TGF-β/Smad signaling pathway. J Mol Histol 2020; 51:77-87. [PMID: 32065356 DOI: 10.1007/s10735-020-09859-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 02/04/2020] [Indexed: 12/15/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a biological process in which tubular epithelial cells lose their phenotypes, and new mesenchymal feature are obtained. In particular, type II EMT possibly contributes to renal tissue fibrogenesis. Recent studies indicate that Lefty-1, a novel member of the TGF-β superfamily with pleiotropical and biological regulation characteristics on TGF-β and other signaling pathways, is considered to have potential fibrotic effects. However, its role in EMT, which is often a long-term consequence of renal tubulointerstitial fibrosis, remains unknown. In this study, we found that Lefty-1 alleviates EMT induction through antagonizing TGF-β/Smad pathway in vivo and in vitro. In unilateral ureteral obstruction (UUO) model mice, administration of adenovirus-mediated overexpression of Lefty-1 (Ad-Lefty-1) significantly reduced TGF-β1/Smad expression and alleviated the phenotypic transition of epithelial cells to mesenchymal cells and extracellular matrix (ECM) accumulation. In high glucose-induced rat renal tubular duct epithelial cell line (NRK-52E), EMT and ECM synthesis were alleviated with Lefty-1 treatment, which significantly inhibited TGF-β1/Smad pathway activation in UUO mice and high glucose-treated NRK-52E cells. Thus, Lefty-1 can alleviate EMT and renal interstitial fibrosis in vivo and in vitro through antagonizing the TGF-β/Smad pathway, and Lefty-1 might have a potential novel therapeutic effect on fibrotic kidney diseases.
Collapse
Affiliation(s)
- Lijun Zhang
- Department of Urology, Minda Hospital, Affiliated to Hubei Minzu University, Enshi, 445000, Hubei, China.
| | - Xiaohua Liu
- Department of Urology, Minda Hospital, Affiliated to Hubei Minzu University, Enshi, 445000, Hubei, China
| | - Jun Liang
- Department of Urology, Minda Hospital, Affiliated to Hubei Minzu University, Enshi, 445000, Hubei, China
| | - Jianhua Wu
- Department of Urology, Minda Hospital, Affiliated to Hubei Minzu University, Enshi, 445000, Hubei, China
| | - Daqing Tan
- Department of Urology, Minda Hospital, Affiliated to Hubei Minzu University, Enshi, 445000, Hubei, China
| | - Wei Hu
- Department of Urology, The First Affiliated Hospital of University of South of China, Hengyang, 421001, Hunan, China
| |
Collapse
|
13
|
De Angelis MT, Santamaria G, Parrotta EI, Scalise S, Lo Conte M, Gasparini S, Ferlazzo E, Aguglia U, Ciampi C, Sgura A, Cuda G. Establishment and characterization of induced pluripotent stem cells (iPSCs) from central nervous system lupus erythematosus. J Cell Mol Med 2019; 23:7382-7394. [PMID: 31536674 PMCID: PMC6815917 DOI: 10.1111/jcmm.14598] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/02/2019] [Accepted: 08/06/2019] [Indexed: 12/16/2022] Open
Abstract
Involvement of the central nervous system (CNS) is an uncommon feature in systemic lupus erythematosus (SLE), making diagnosis rather difficult and challenging due to the poor specificity of neuropathic symptoms and neurological symptoms. In this work, we used human‐induced pluripotent stem cells (hiPSCs) derived from CNS‐SLE patient, with the aim to dissect the molecular insights underlying the disease by gene expression analysis and modulation of implicated pathways. CNS‐SLE‐derived hiPSCs allowed us to provide evidence of Erk and Akt pathways involvement and to identify a novel cohort of potential biomarkers, namely CHCHD2, IDO1, S100A10, EPHA4 and LEFTY1, never reported so far. We further extended the study analysing a panel of oxidative stress‐related miRNAs and demonstrated, under normal or stress conditions, a strong dysregulation of several miRNAs in CNS‐SLE‐derived compared to control hiPSCs. In conclusion, we provide evidence that iPSCs reprogrammed from CNS‐SLE patient are a powerful useful tool to investigate the molecular mechanisms underlying the disease and to eventually develop innovative therapeutic approaches.
Collapse
Affiliation(s)
- Maria Teresa De Angelis
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Gianluca Santamaria
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Elvira Immacolata Parrotta
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Stefania Scalise
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Michela Lo Conte
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Sara Gasparini
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Edoardo Ferlazzo
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy.,Regional Epilepsy Centre, Great Metropolitan Hospital, Reggio Calabria, Italy
| | - Umberto Aguglia
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy.,Regional Epilepsy Centre, Great Metropolitan Hospital, Reggio Calabria, Italy
| | - Clara Ciampi
- Department of Science, University of Rome " Roma Tre", Rome, Italy
| | - Antonella Sgura
- Department of Science, University of Rome " Roma Tre", Rome, Italy
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| |
Collapse
|
14
|
Sandomenico A, Ruvo M. Targeting Nodal and Cripto-1: Perspectives Inside Dual Potential Theranostic Cancer Biomarkers. Curr Med Chem 2019; 26:1994-2050. [PMID: 30207211 DOI: 10.2174/0929867325666180912104707] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Elucidating the mechanisms of recurrence of embryonic signaling pathways in tumorigenesis has led to the discovery of onco-fetal players which have physiological roles during normal development but result aberrantly re-activated in tumors. In this context, Nodal and Cripto-1 are recognized as onco-developmental factors, which are absent in normal tissues but are overexpressed in several solid tumors where they can serve as theranostic agents. OBJECTIVE To collect, review and discuss the most relevant papers related to the involvement of Nodal and Cripto-1 in the development, progression, recurrence and metastasis of several tumors where they are over-expressed, with a particular attention to their occurrence on the surface of the corresponding sub-populations of cancer stem cells (CSC). RESULTS We have gathered, rationalized and discussed the most interesting findings extracted from some 370 papers related to the involvement of Cripto-1 and Nodal in all tumor types where they have been detected. Data demonstrate the clear connection between Nodal and Cripto-1 presence and their multiple oncogenic activities across different tumors. We have also reviewed and highlighted the potential of targeting Nodal, Cripto-1 and the complexes that they form on the surface of tumor cells, especially of CSC, as an innovative approach to detect and suppress tumors with molecules that block one or more mechanisms that they regulate. CONCLUSION Overall, Nodal and Cripto-1 represent two innovative and effective biomarkers for developing potential theranostic anti-tumor agents that target normal as well as CSC subpopulations and overcome both pharmacological resistance and tumor relapse.
Collapse
Affiliation(s)
- Annamaria Sandomenico
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche (IBB-CNR), via Mezzocannone, 16, 80134, Napoli, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche (IBB-CNR), via Mezzocannone, 16, 80134, Napoli, Italy
| |
Collapse
|
15
|
Decoding epigenetic cell signaling in neuronal differentiation. Semin Cell Dev Biol 2019; 95:12-24. [PMID: 30578863 DOI: 10.1016/j.semcdb.2018.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/18/2018] [Indexed: 12/18/2022]
Abstract
Neurogenesis is the process by which new neurons are generated in the brain. Neural stem cells (NSCs) are differentiated into neurons, which are integrated into the neural network. Nowadays, pluripotent stem cells, multipotent stem cells, and induced pluripotent stem cells can be artificially differentiated into neurons utilizing several techniques. Specific transcriptional profiles from NSCs during differentiation are frequently used to approach and observe phenotype alteration and functional determination of neurons. In this context, the role of non-coding RNA, transcription factors and epigenetic changes in neuronal development and differentiation has gained importance. Epigenetic elucidation has become a field of intense research due to distinct patterns of normal conditions and different neurodegenerative disorders, which can be explored to develop new diagnostic methods or gene therapies. In this review, we discuss the complexity of transcription factors, non-coding RNAs, and extracellular vesicles that are responsible for guiding and coordinating neural development.
Collapse
|
16
|
Li H, Cao R, Bai L, Qiao XM, Zhao YQ. Lefty promotes the proliferation and invasion of trophoblast cells by inhibiting nodal expression. Cell Biol Int 2018; 42:1259-1264. [PMID: 29663570 DOI: 10.1002/cbin.10976] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 04/06/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Hong Li
- Obstetric Department; The Maternity Hospital of DaLian; Dalian 116033 China
| | - Rui Cao
- Gynecology Department; The Maternity Hospital of DaLian; Dalian 116033 China
| | - Liang Bai
- Oral Maxillofacial Surgery Department; The First Affiliated Hospital of DaLian Medical University; Dalian 116023 China
| | - Xin-min Qiao
- Obstetric Department; The Maternity Hospital of DaLian; Dalian 116033 China
| | - Yong-qin Zhao
- Obstetric Department; The Maternity Hospital of DaLian; Dalian 116033 China
| |
Collapse
|
17
|
TGF-β-mediated LEFTY/Akt/GSK-3β/Snail axis modulates epithelial-mesenchymal transition and cancer stem cell properties in ovarian clear cell carcinomas. Mol Carcinog 2018; 57:957-967. [DOI: 10.1002/mc.22816] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 03/19/2018] [Accepted: 03/27/2018] [Indexed: 01/05/2023]
|
18
|
Kalyan A, Carneiro BA, Chandra S, Kaplan J, Chae YK, Matsangou M, Hendrix MJC, Giles F. Nodal Signaling as a Developmental Therapeutics Target in Oncology. Mol Cancer Ther 2018; 16:787-792. [PMID: 28468864 DOI: 10.1158/1535-7163.mct-16-0215] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 11/22/2016] [Accepted: 12/05/2016] [Indexed: 11/16/2022]
Abstract
The tumor microenvironment is a vital feature of oncogenesis and tumor progression. There are several parallels between cancer cells and early developmental stem cells, including their plasticity and signaling mechanisms. In early fetal development, Nodal is expressed for endodermal and mesodermal differentiation. This expression has been shown reemerge in the setting of epithelial cancers, such as breast and melanoma. High Nodal expression correlates to an aggressive tumor grade in these malignancies. Nodal signal begins with its interaction with its coreceptor, Cripto-1, leading to activation of Smad2/Smad3 and ultimately downstream transcription and translation. Lefty is the natural inhibitor of Nodal and controls Nodal signaling during fetal development. However, cancer cells lack the presence of Lefty, thus leading to uncontrolled tumor growth. Given this understanding, inhibition of the Nodal pathway offers a new novel therapeutic target in oncology. Mol Cancer Ther; 16(5); 787-92. ©2017 AACR.
Collapse
Affiliation(s)
- Aparna Kalyan
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois. .,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Benedito A Carneiro
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Sunandana Chandra
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Jason Kaplan
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Young Kwang Chae
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Maria Matsangou
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Mary J C Hendrix
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois.,Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Anne and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Francis Giles
- Developmental Therapeutics Program, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Olson Pavilion, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| |
Collapse
|
19
|
Fei W, Kijima D, Hashimoto M, Hashimura M, Oguri Y, Kajita S, Matsumoto T, Yokoi A, Saegusa M. A functional role of LEFTY during progesterone therapy for endometrial carcinoma. Cell Commun Signal 2017; 15:56. [PMID: 29268772 PMCID: PMC5740891 DOI: 10.1186/s12964-017-0211-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/13/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The left-right determination factor (LEFTY) is a novel member of the TGF-β/Smad2 pathway and belongs to the premenstrual/menstrual repertoire in human endometrium, but little is known about its functional role in endometrial carcinomas (Em Cas). Herein, we focused on LEFTY expression and its association with progesterone therapy in Em Cas. METHODS Regulation and function of LEFTY, as well as its associated molecules including Smad2, ovarian hormone receptors, GSK-3β, and cell cycle-related factors, were assessed using clinical samples and cell lines of Em Cas. RESULTS In clinical samples, LEFTY expression was positively correlated with estrogen receptor-α, but not progesterone receptor (PR), status, and was inversely related to phosphorylated (p) Smad2, cyclin A2, and Ki-67 levels. During progesterone therapy, expression of LEFTY, pSmad2, and pGSK-3β showed stepwise increases, with significant correlations to morphological changes toward secretory features and decreased Ki-67 values. In Ishikawa cells, an Em Ca cell line that expresses PR, progesterone treatment reduced proliferation and induced increased expression of LEFTY and pGSK-3β, although LEFTY promoter regions were inhibited by transfection of PR. Moreover, inhibition of GSK-3β resulted in increased LEFTY expression through a decrease in its ubiquitinated form, suggesting posttranslational regulation of LEFTY protein via GSK-3β suppression in response to progesterone. In addition, overexpression or knockdown of LEFTY led to suppression or enhancement of Smad2-dependent cyclin A2 expression, respectively. CONCLUSION Upregulation of LEFTY may serve as a useful clinical marker for the therapeutic effects of progesterone for Em Cas, leading to inhibition of tumor cell proliferation through alteration in Smad2-dependent transcription of cyclin A2.
Collapse
Affiliation(s)
- Wu Fei
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.,Department of Gynecology and Obstetrics, Jilin University Bethune Second Hospital, Changchun, People's Republic of China
| | - Daiki Kijima
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Mami Hashimoto
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Miki Hashimura
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yasuko Oguri
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Sabine Kajita
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Toshihide Matsumoto
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Ako Yokoi
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Makoto Saegusa
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.
| |
Collapse
|
20
|
Zhang L, Xu C, Hu W, Wu P, Qin C, Zhang J. Anti-inflammatory effects of Lefty-1 in renal tubulointerstitial inflammation via regulation of the NF-κB pathway. Int J Mol Med 2017; 41:1293-1304. [PMID: 29286065 PMCID: PMC5819905 DOI: 10.3892/ijmm.2017.3327] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 11/23/2017] [Indexed: 11/13/2022] Open
Abstract
Renal tubulointerstitial inflammation has an important role in fibrosis, which is the main pathogenetic alteration associated with chronic kidney disease (CKD). The left-right determination factor 1 (Lefty-1) gene pleiotropically and biologically regulates transforming growth factor, mitogen-activated protein kinase and other signaling pathways, and is considered to have a potential anti-inflammatory function. However, its role in renal tubulointerstitial inflammation, which is often a long-term consequence of renal fibrosis, is currently unknown. In the present study, the effects of adenovirus-mediated overexpression of Lefty-1 (Ad-Lefty-1-flag) on renal tubulointerstitial inflammation were determined using a mouse model of unilateral ureteral obstruction (UUO) and a rat renal tubular duct epithelial cell line (NRK-52E), which was treated with lipopolysaccharide (LPS). In vivo results indicated that the inflammatory response was increased in UUO mice, as evidenced by the increase in inflammatory cytokines and chemokines. Conversely, Lefty-1 significantly reversed the effects of UUO. Furthermore, the results of the in vitro study demonstrated that Lefty-1 significantly inhibited LPS-induced inflammatory marker expression in cultured NRK-52E cells via the nuclear factor (NF)-κB signaling pathway. These results suggested that Lefty-1 may ameliorate renal tubulointerstitial inflammation by suppressing NF-κB signaling. In conclusion, the findings of the present study indicated that Lefty-1 may be considered a potential novel therapeutic agent for inhibiting renal tubulointerstitial inflammation or even reversing the CKD process.
Collapse
Affiliation(s)
- Lijun Zhang
- Department of Urology, Minda Hospital Affiliated to Hubei Institute for Nationalities, Enshi, Hubei 445000, P.R. China
| | - Changgeng Xu
- Department of Urology, Wuhan Central Hospital, Wuhan, Hubei 430014, P.R. China
| | - Wei Hu
- Department of Urology, The First Affiliated Hospital of University of South of China, Hengyang, Hunan 421001, P.R. China
| | - Pin Wu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Cong Qin
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jie Zhang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
21
|
LEFTY2 inhibits endometrial receptivity by downregulating Orai1 expression and store-operated Ca 2+ entry. J Mol Med (Berl) 2017; 96:173-182. [PMID: 29230527 PMCID: PMC5778154 DOI: 10.1007/s00109-017-1610-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 10/16/2017] [Accepted: 11/02/2017] [Indexed: 12/16/2022]
Abstract
Abstract Early embryo development and endometrial differentiation are initially independent processes, and synchronization, imposed by a limited window of implantation, is critical for reproductive success. A putative negative regulator of endometrial receptivity is LEFTY2, a member of the transforming growth factor (TGF)-β family. LEFTY2 is highly expressed in decidualizing human endometrial stromal cells (HESCs) during the late luteal phase of the menstrual cycle, coinciding with the closure of the window of implantation. Here, we show that flushing of the uterine lumen in mice with recombinant LEFTY2 inhibits the expression of key receptivity genes, including Cox2, Bmp2, and Wnt4, and blocks embryo implantation. In Ishikawa cells, a human endometrial epithelial cell line, LEFTY2 downregulated the expression of calcium release-activated calcium channel protein 1, encoded by ORAI1, and inhibited store-operated Ca2+ entry (SOCE). Furthermore, LEFTY2 and the Orai1 blockers 2-APB, MRS-1845, as well as YM-58483, inhibited, whereas the Ca2+ ionophore, ionomycin, strongly upregulated COX2, BMP2 and WNT4 expression in decidualizing HESCs. These findings suggest that LEFTY2 closes the implantation window, at least in part, by downregulating Orai1, which in turn limits SOCE and antagonizes expression of Ca2+-sensitive receptivity genes. Key messages •Endometrial receptivity is negatively regulated by LEFTY2. •LEFTY2 inhibits the expression of key murine receptivity genes, including Cox2, Bmp2and Wnt4, and blocks embryo implantation. •LEFTY2 downregulates the expression of Orai1 and inhibits SOCE. •LEFTY2 and the Orai1 blockers 2-APB, MRS-1845, and YM-58483 inhibit COX2, BMP2, and WNT4 expression in endometrial cells. •Targeting LEFTY2 and Orai1 may represent a novel approach for treating unexplained infertility. Electronic supplementary material The online version of this article (10.1007/s00109-017-1610-9) contains supplementary material, which is available to authorized users.
Collapse
|
22
|
Whirledge S, Kisanga EP, Taylor RN, Cidlowski JA. Pioneer Factors FOXA1 and FOXA2 Assist Selective Glucocorticoid Receptor Signaling in Human Endometrial Cells. Endocrinology 2017; 158:4076-4092. [PMID: 28938408 PMCID: PMC5695839 DOI: 10.1210/en.2017-00361] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 08/24/2017] [Indexed: 01/10/2023]
Abstract
Successful pregnancy relies on dynamic control of cell signaling to achieve uterine receptivity and the necessary biological changes required for endometrial decidualization, embryo implantation, and fetal development. Glucocorticoids are master regulators of intracellular signaling and can directly regulate embryo implantation and endometrial remodeling during murine pregnancy. In immortalized human uterine cells, we have shown that glucocorticoids and estradiol (E2) coregulate thousands of genes. Recently, glucocorticoids and E2 were shown to coregulate the expression of Left-right determination factor 1 (LEFTY1), previously implicated in the regulation of decidualization. To elucidate the molecular mechanism by which glucocorticoids and E2 regulate the expression of LEFTY1, immortalized and primary human endometrial cells were evaluated for gene expression and receptor recruitment to regulatory regions of the LEFTY1 gene. Glucocorticoid administration induced expression of LEFTY1 messenger RNA and protein and recruitment of the glucocorticoid receptor (GR) and activated polymerase 2 to the promoter of LEFTY1. Glucocorticoid-mediated recruitment of GR was dependent on pioneer factors FOXA1 and FOXA2. E2 was found to antagonize glucocorticoid-mediated induction of LEFTY1 by reducing recruitment of GR, FOXA1, FOXA2, and activated polymerase 2 to the LEFTY1 promoter. Gene expression analysis identified several genes whose glucocorticoid-dependent induction required FOXA1 and FOXA2 in endometrial cells. These results suggest a molecular mechanism by which E2 antagonizes GR-dependent induction of specific genes by preventing the recruitment of the pioneer factors FOXA1 and FOXA2 in a physiologically relevant model.
Collapse
Affiliation(s)
- Shannon Whirledge
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut 06520
| | - Edwina P. Kisanga
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut 06520
| | - Robert N. Taylor
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27101
| | - John A. Cidlowski
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709
| |
Collapse
|
23
|
Akiya M, Yamazaki M, Matsumoto T, Kawashima Y, Oguri Y, Kajita S, Kijima D, Chiba R, Yokoi A, Takahashi H, Kodera Y, Saegusa M. Identification of LEFTY as a molecular marker for ovarian clear cell carcinoma. Oncotarget 2017; 8:63646-63664. [PMID: 28969018 PMCID: PMC5609950 DOI: 10.18632/oncotarget.18882] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 06/11/2017] [Indexed: 01/16/2023] Open
Abstract
To identify proteins involved in ovarian clear cell carcinoma (OCCCa), shotgun proteomics analysis was applied using formalin-fixed and paraffin-embedded samples of ovarian carcinoma. Analysis of 1521 proteins revealed that 52 were differentially expressed between four OCCCa and 12 non-OCCCa samples. Of the highly expressed proteins in OCCCa, we focused on left-right determination factor (LEFTY), a novel member of the transforming growth factor-β superfamily. In 143 cases of ovarian epithelial carcinoma including 99 OCCCas and 44 non-OCCCas, LEFTY expression at both mRNA and protein levels was significantly higher in OCCCas compared with non-OCCCas, with the mRNA expression of LEFTY1 being predominant compared to that of LEFTY2. OCCCa cells stably overexpressing LEFTY1 showed reduced cell proliferation, along with decreased pSmad2 expression, and also either displayed an activated p53/p21waf1 pathway or increased p27kip1 expression, directly or indirectly. Moreover, the treatment of stable cell lines with cisplatin led to increased apoptotic cells, together with the inhibition of protein expression of a pSmad2-mediated X-linked inhibitor of apoptosis and a decreased bcl2/bax ratio. Blocking LEFTY1 expression with a specific short hairpin RNA inhibited cisplatin-induced apoptosis, probably through the increased expression of both XIAP and bcl2, but not bax. In clinical samples, a significantly higher number of apoptotic cells and lower Ki-67 labeling indices were observed in OCCCas with a high LEFTY score relative to those with a low score. These findings suggest that LEFTY may be an excellent OCCCa-specific molecular marker, which has anti-tumor effects in altering cell proliferation and cellular susceptibility to apoptosis.
Collapse
Affiliation(s)
- Masashi Akiya
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Masaaki Yamazaki
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Toshihide Matsumoto
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Yusuke Kawashima
- Center for Disease Proteomics, School of Science, Kitasato University, Sagamihara, Kanagawa 252-0374, Japan
| | - Yasuko Oguri
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Sabine Kajita
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Daiki Kijima
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Risako Chiba
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Ako Yokoi
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Hiroyuki Takahashi
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| | - Yoshio Kodera
- Center for Disease Proteomics, School of Science, Kitasato University, Sagamihara, Kanagawa 252-0374, Japan
| | - Makoto Saegusa
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa 252-0374, Japan
| |
Collapse
|
24
|
Ma H, Qiao S, Wang Z, Geng S, Zhao Y, Hou X, Tian W, Chen X, Yao L. Microencapsulation of Lefty-secreting engineered cells for pulmonary fibrosis therapy in mice. Am J Physiol Lung Cell Mol Physiol 2017; 312:L741-L747. [PMID: 28213468 DOI: 10.1152/ajplung.00295.2016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 02/08/2017] [Accepted: 02/08/2017] [Indexed: 11/22/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease that causes unremitting deposition of extracellular matrix proteins, thus resulting in distortion of the pulmonary architecture and impaired gas exchange. Associated with high morbidity and mortality, IPF is generally refractory to current pharmacological therapies. Lefty A, a potent inhibitor of transforming growth factor-β signaling, has been shown to have promising antifibrotic ability in vitro for the treatment of renal fibrosis and other potential organ fibroses. Here, we determined whether Lefty A can attenuate bleomycin (BLM)-induced pulmonary fibrosis in vivo based on a novel therapeutic strategy where human embryonic kidney 293 (HEK293) cells are genetically engineered with the Lefty A-associated GFP gene. The engineered HEK293 cells were encapsulated in alginate microcapsules and then subcutaneously implanted in ICR mice that had 1 wk earlier been intratracheally administered BLM to induce pulmonary fibrosis. The severity of fibrosis in lung tissue was assessed using pathological morphology and collagen expression to examine the effect of Lefty A released from the microencapsulated cells. The engineered HEK293 cells with Lefty A significantly reduced the expression of connective tissue growth factor and collagen type I mRNA, lessened the morphological fibrotic effects induced by BLM, and increased the expression of matrix metalloproteinase-9. This illustrates that engineered HEK293 cells with Lefty A can attenuate pulmonary fibrosis in vivo, thus providing a novel method to treat human pulmonary fibrotic disease and other organ fibroses.
Collapse
Affiliation(s)
- Hongge Ma
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shupei Qiao
- Bio-X Center, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Zeli Wang
- Bio-X Center, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Shuai Geng
- Department of Pharmacology, Harbin Medical University, Harbin, China; and
| | - Yufang Zhao
- Bio-X Center, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xiaolu Hou
- Bio-X Center, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Weiming Tian
- Bio-X Center, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xiongbiao Chen
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Lifen Yao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China;
| |
Collapse
|
25
|
Crespo D, Assis LHC, Furmanek T, Bogerd J, Schulz RW. Expression profiling identifies Sertoli and Leydig cell genes as Fsh targets in adult zebrafish testis. Mol Cell Endocrinol 2016; 437:237-251. [PMID: 27566230 DOI: 10.1016/j.mce.2016.08.033] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/27/2016] [Accepted: 08/22/2016] [Indexed: 11/26/2022]
Abstract
Spermatogonial stem cells are quiescent, undergo self-renewal or differentiating divisions, thereby forming the cellular basis of spermatogenesis. This cellular development is orchestrated by follicle-stimulating hormone (FSH), through the production of Sertoli cell-derived factors, and by Leydig cell-released androgens. Here, we investigate the transcriptional events induced by Fsh in a steroid-independent manner on the restart of zebrafish (Danio rerio) spermatogenesis ex vivo, using testis from adult males where type A spermatogonia were enriched by estrogen treatment in vivo. Under these conditions, RNA sequencing preferentially detected differentially expressed genes in somatic/Sertoli cells. Fsh-stimulated spermatogonial proliferation was accompanied by modulating several signaling systems (i.e. Tgf-β, Hedgehog, Wnt and Notch pathways). In silico protein-protein interaction analysis indicated a role for Hedgehog family members potentially integrating signals from different pathways during fish spermatogenesis. Moreover, Fsh had a marked impact on metabolic genes, such as lactate and fatty acid metabolism, or on Sertoli cell barrier components. Fish Leydig cells express the Fsh receptor and one of the most robust Fsh-responsive genes was insulin-like 3 (insl3), a Leydig cell-derived growth factor. Follow-up work showed that recombinant zebrafish Insl3 mediated pro-differentiation effects of Fsh on spermatogonia in an androgen-independent manner. Our experimental approach allowed focusing on testicular somatic genes in zebrafish and showed that the activity of signaling systems known to be relevant in stem cells was modulated by Fsh, providing promising leads for future work, as exemplified by the studies on Insl3.
Collapse
Affiliation(s)
- Diego Crespo
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Luiz H C Assis
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Tomasz Furmanek
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Jan Bogerd
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Rüdiger W Schulz
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands; Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway.
| |
Collapse
|
26
|
Li X, Shi S, Li FF, Cheng R, Han Y, Diao LW, Zhang Q, Zhi JX, Liu SL. Characterization of soluble N-ethylmaleimide-sensitive factor attachment protein receptor gene STX18 variations for possible roles in congenital heart diseases. Gene 2016; 598:79-83. [PMID: 27816473 DOI: 10.1016/j.gene.2016.10.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/19/2016] [Accepted: 10/31/2016] [Indexed: 02/07/2023]
Abstract
Congenital heart disease (CHD) is among the most prevalent and complex congenital anatomic malformations in newborns. Interactions of cardiac progenitor with a broad range of cellular regulatory factors play key roles in the formation of mammalian heart and pathogenesis of CHD. STX18 is a soluble N-ethylmaleimide-sensitive factor attachment protein receptor, which is involved in numeral cellular activities such as organelle assembly and the cell cycle. The aim of this work was to find evidence on whether STX18 variations might be associated with CHD in Chinese Han populations. We evaluated SNPs rs2044, rs33952588, rs61740788, rs12504020 and rs12644497, which are located within the exon or intron sequences of the STX18 gene, for 310 Chinese Han CHD patients and 400 non-CHD controls. Using SPSS software (version 19.0) and the online software OEGE, we conducted statistical analyses and Hardy-Weinberg equilibrium test, respectively. Among the five SNPs identified in the STX18 gene, rs33952588 and rs61740788 had very low genetic heterozygosity. In contrast, the genetic heterozygosity of the remaining three variations rs12504020 and rs12644497 near the 5'UTR and rs2044 within 3'UTR of the STX18 gene was considerably high. Analysis of associations of these genetic variations with the risk of CHD showed that rs12644497 (P value=0.017<0.05) was associated with the risk of CHD, specifically VSD and ASD, whereas rs12504020 (P value=0.560>0.05) and rs2044 (P value=0.972>0.05) were not. The SNP rs12644497 in the STX18 gene was associated with CHD in Chinese Han populations.
Collapse
Affiliation(s)
- Xia Li
- Systemomics Center, College of Pharmacy, and Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Shuai Shi
- Department of Cardiology of the Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fei-Feng Li
- Systemomics Center, College of Pharmacy, and Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China.
| | - Rui Cheng
- Department of Cardiology of the Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ying Han
- Department of Cardiology of the Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Li-Wei Diao
- Department of Cardiology of the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiong Zhang
- Department of Antibiotics, Heilongjiang Province Food and Drug Inspection Testing Institute, Harbin, China
| | - Ji-Xin Zhi
- Department of Cardiology of the Fourth Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Shu-Lin Liu
- Systemomics Center, College of Pharmacy, and Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China; Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Canada.
| |
Collapse
|
27
|
Quantitative Profiling of Single Formalin Fixed Tumour Sections: proteomics for translational research. Sci Rep 2016; 6:34949. [PMID: 27713570 PMCID: PMC5054533 DOI: 10.1038/srep34949] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/20/2016] [Indexed: 02/06/2023] Open
Abstract
Although re-sequencing of gene panels and mRNA expression profiling are now firmly established in clinical laboratories, in-depth proteome analysis has remained a niche technology, better suited for studying model systems rather than challenging materials such as clinical trial samples. To address this limitation, we have developed a novel and optimized platform called SP3-Clinical Tissue Proteomics (SP3-CTP) for in-depth proteome profiling of practical quantities of tumour tissues, including formalin fixed and paraffin embedded (FFPE). Using single 10 μm scrolls of clinical tumour blocks, we performed in-depth quantitative analyses of individual sections from ovarian tumours covering the high-grade serous, clear cell, and endometrioid histotypes. This examination enabled the generation of a novel high-resolution proteome map of ovarian cancer histotypes from clinical tissues. Comparison of the obtained proteome data with large-scale genome and transcriptome analyses validated the observed proteome biology for previously validated hallmarks of this disease, and also identified novel protein features. A tissue microarray analysis validated cystathionine gamma-lyase (CTH) as a novel clear cell carcinoma feature with potential clinical relevance. In addition to providing a milestone in the understanding of ovarian cancer biology, these results show that in-depth proteomic analysis of clinically annotated FFPE materials can be effectively used as a biomarker discovery tool and perhaps ultimately as a diagnostic approach.
Collapse
|
28
|
Khalkhali-Ellis Z, Galat V, Galat Y, Gilgur A, Seftor EA, Hendrix MJC. Lefty Glycoproteins in Human Embryonic Stem Cells: Extracellular Delivery Route and Posttranslational Modification in Differentiation. Stem Cells Dev 2016; 25:1681-1690. [PMID: 27554431 DOI: 10.1089/scd.2016.0081] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Lefty is a member of transforming growth factor-beta (TGF-β) superfamily and a potent antagonist of the TGF-β/Nodal/Activin signaling pathway. Lefty is critical in sustaining self-renewal/pluripotency status, and implicated in the differentiation of embryonic stem cells (ESCs). However, emerging studies depict Lefty as a multifaceted protein involved in myriad cellular events. Lefty proteins (human Lefty A and B) are secreted glycoproteins, but their mode of secretion and the significance of their "glycan" moiety remain mostly unexplored. By employing an in vitro system of human ESCs (hESCs), we observed that Lefty protein(s) are encased in exosomes for extracellular release. The exosomal- and cell-associated Lefty diverge in their proteolytic processing, and possess N-glycan structures of high mannose and complex nature. Differentiation of hESCs to mesenchymal cells (MSCs) or neuronal progenitor cells (NPCs) entails distinct changes in the Lefty A/Lefty B gene(s), and protein expression. Specifically, the proteolytic cleavage and N-glycan composition of the cell-associated and exosomal Lefty differ in the differentiated progenies. These modifications affected Lefty's inhibitory effect on Nodal signaling in aggressive melanoma cells. The microheterogeneity in the processing and glycosylation of Lefty protein(s) between hESCs, MSCs, and NPCs could present efficient means of diversifying the endogenous functions of Lefty. Whether Lefty's diverse functions in embryonic patterning, as well as its diffusion range in the extracellular environment, are similarly affected remains to be determined. Our studies underscore the potential relevance of Lefty-packaged exosomes for combating debilitating diseases such as cancer.
Collapse
Affiliation(s)
- Zhila Khalkhali-Ellis
- 1 Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute , Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Vasiliy Galat
- 2 Department of Pathology, Stanley Manne Children's Research Institute , Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,3 Developmental Biology Program, Stanley Manne Children's Research Institute , Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,4 Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine , Chicago, Illinois
| | - Yekaterina Galat
- 3 Developmental Biology Program, Stanley Manne Children's Research Institute , Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Alina Gilgur
- 1 Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute , Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Elisabeth A Seftor
- 1 Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute , Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Mary J C Hendrix
- 1 Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute , Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,4 Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine , Chicago, Illinois
| |
Collapse
|
29
|
LeftyA decreases Actin Polymerization and Stiffness in Human Endometrial Cancer Cells. Sci Rep 2016; 6:29370. [PMID: 27404958 PMCID: PMC4941646 DOI: 10.1038/srep29370] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 06/16/2016] [Indexed: 12/27/2022] Open
Abstract
LeftyA, a cytokine regulating stemness and embryonic differentiation, down-regulates cell proliferation and migration. Cell proliferation and motility require actin reorganization, which is under control of ras-related C3 botulinum toxin substrate 1 (Rac1) and p21 protein-activated kinase 1 (PAK1). The present study explored whether LeftyA modifies actin cytoskeleton, shape and stiffness of Ishikawa cells, a well differentiated endometrial carcinoma cell line. The effect of LeftyA on globular over filamentous actin ratio was determined utilizing Western blotting and flow cytometry. Rac1 and PAK1 transcript levels were measured by qRT-PCR as well as active Rac1 and PAK1 by immunoblotting. Cell stiffness (quantified by the elastic modulus), cell surface area and cell volume were studied by atomic force microscopy (AFM). As a result, 2 hours treatment with LeftyA (25 ng/ml) significantly decreased Rac1 and PAK1 transcript levels and activity, depolymerized actin, and decreased cell stiffness, surface area and volume. The effect of LeftyA on actin polymerization was mimicked by pharmacological inhibition of Rac1 and PAK1. In the presence of the Rac1 or PAK1 inhibitor LeftyA did not lead to significant further actin depolymerization. In conclusion, LeftyA leads to disruption of Rac1 and Pak1 activity with subsequent actin depolymerization, cell softening and cell shrinkage.
Collapse
|
30
|
Morikawa M, Derynck R, Miyazono K. TGF-β and the TGF-β Family: Context-Dependent Roles in Cell and Tissue Physiology. Cold Spring Harb Perspect Biol 2016; 8:8/5/a021873. [PMID: 27141051 DOI: 10.1101/cshperspect.a021873] [Citation(s) in RCA: 955] [Impact Index Per Article: 106.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The transforming growth factor-β (TGF-β) is the prototype of the TGF-β family of growth and differentiation factors, which is encoded by 33 genes in mammals and comprises homo- and heterodimers. This review introduces the reader to the TGF-β family with its complexity of names and biological activities. It also introduces TGF-β as the best-studied factor among the TGF-β family proteins, with its diversity of roles in the control of cell proliferation and differentiation, wound healing and immune system, and its key roles in pathology, for example, skeletal diseases, fibrosis, and cancer.
Collapse
Affiliation(s)
- Masato Morikawa
- Ludwig Cancer Research, Science for Life Laboratory, Uppsala University, Biomedical Center, SE-751 24 Uppsala, Sweden
| | - Rik Derynck
- Department of Cell and Tissue Biology, University of California at San Francisco, San Francisco, California 94143
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
31
|
Lee YJ, Ramakrishna S, Chauhan H, Park WS, Hong SH, Kim KS. Dissecting microRNA-mediated regulation of stemness, reprogramming, and pluripotency. ACTA ACUST UNITED AC 2016; 5:2. [PMID: 27006752 PMCID: PMC4802578 DOI: 10.1186/s13619-016-0028-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 02/20/2016] [Indexed: 02/06/2023]
Abstract
Increasing evidence indicates that microRNAs (miRNAs), endogenous short non-coding RNAs 19–24 nucleotides in length, play key regulatory roles in various biological events at the post-transcriptional level. Embryonic stem cells (ESCs) represent a valuable tool for disease modeling, drug discovery, developmental studies, and potential cell-based therapies in regenerative medicine due to their unlimited self-renewal and pluripotency. Therefore, remarkable progress has been made in recent decades toward understanding the expression and functions of specific miRNAs in the establishment and maintenance of pluripotency. Here, we summarize the recent knowledge regarding the regulatory roles of miRNAs in self-renewal of pluripotent ESCs and during cellular reprogramming, as well as the potential role of miRNAs in two distinct pluripotent states (naïve and primed).
Collapse
Affiliation(s)
- Young Jin Lee
- iDream Research Center, MizMedi Women's Hospital, Seoul, 07639 South Korea
| | - Suresh Ramakrishna
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763 South Korea.,College of Medicine, Hanyang University, Seoul, South Korea
| | | | - Won Sun Park
- Department of Physiology, School of Medicine, Kangwon National University, Chuncheon, 24341 South Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Gangwon-do 24341 South Korea.,Stem Cell Institute, Kangwon National University, Chuncheon, 24341 South Korea
| | - Kye-Seong Kim
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763 South Korea.,College of Medicine, Hanyang University, Seoul, South Korea
| |
Collapse
|
32
|
Terry S, El-Sayed IY, Destouches D, Maillé P, Nicolaiew N, Ploussard G, Semprez F, Pimpie C, Beltran H, Londono-Vallejo A, Allory Y, de la Taille A, Salomon DS, Vacherot F. CRIPTO overexpression promotes mesenchymal differentiation in prostate carcinoma cells through parallel regulation of AKT and FGFR activities. Oncotarget 2016; 6:11994-2008. [PMID: 25596738 PMCID: PMC4494918 DOI: 10.18632/oncotarget.2740] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 11/11/2014] [Indexed: 02/03/2023] Open
Abstract
Members of the EGF-CFC (Cripto, FRL-1, Cryptic) protein family are increasingly recognized as key mediators of cell movement and cell differentiation during vertebrate embryogenesis. The founding member of this protein family, CRIPTO, is overexpressed in various human carcinomas. Yet, the biological role of CRIPTO in this setting remains unclear. Here, we find CRIPTO expression as especially high in a subgroup of primary prostate carcinomas with poorer outcome, wherein resides cancer cell clones with mesenchymal traits. Experimental studies in PCa models showed that one notable function of CRIPTO expression in prostate carcinoma cells may be to augment PI3K/AKT and FGFR1 signaling, which promotes epithelial-mesenchymal transition and sustains a mesenchymal state. In the observed signaling events, FGFR1 appears to function parallel to AKT, and the two pathways act cooperatively to enhance migratory, invasive and transformation properties specifically in the CRIPTO overexpressing cells. Collectively, these findings suggest a novel molecular network, involving CRIPTO, AKT, and FGFR signaling, in favor of the emergence of mesenchymal-like cancer cells during the development of aggressive prostate tumors.
Collapse
Affiliation(s)
- Stéphane Terry
- Inserm, U955, Equipe 7, Créteil, France.,Université Paris-Est, UMR_S955, UPEC, Créteil, France.,Institut Curie, Centre de Recherche, CNRS UMR 3244, Paris, France.,Inserm, U753, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - Ihsan Y El-Sayed
- Inserm, U955, Equipe 7, Créteil, France.,Université Paris-Est, UMR_S955, UPEC, Créteil, France.,EDST/PRASE, Rafic Harriri Campus, Faculté des Sciences, Université Libanaise, Beyrouth, Liban
| | - Damien Destouches
- Inserm, U955, Equipe 7, Créteil, France.,Université Paris-Est, UMR_S955, UPEC, Créteil, France.,Laboratoire de Recherche sur la Croissance Cellulaire, la Réparation et la Régénération Tissulaires (CRRET), CNRS, Créteil, France
| | - Pascale Maillé
- AP-HP, Hôpital H. Mondor, Département de Pathologie, Créteil, France
| | - Nathalie Nicolaiew
- Inserm, U955, Equipe 7, Créteil, France.,Université Paris-Est, UMR_S955, UPEC, Créteil, France
| | - Guillaume Ploussard
- Inserm, U955, Equipe 7, Créteil, France.,AP-HP, Hôpital H. Mondor, Service d'urologie, Créteil, France
| | - Fannie Semprez
- Inserm, U955, Equipe 7, Créteil, France.,Université Paris-Est, UMR_S955, UPEC, Créteil, France
| | - Cynthia Pimpie
- Inserm, U955, Equipe 7, Créteil, France.,Université Paris-Est, UMR_S955, UPEC, Créteil, France
| | - Himisha Beltran
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | - Yves Allory
- Inserm, U955, Equipe 7, Créteil, France.,Université Paris-Est, UMR_S955, UPEC, Créteil, France.,AP-HP, Hôpital H. Mondor, Département de Pathologie, Créteil, France
| | - Alexandre de la Taille
- Inserm, U955, Equipe 7, Créteil, France.,Université Paris-Est, UMR_S955, UPEC, Créteil, France.,AP-HP, Hôpital H. Mondor, Service d'urologie, Créteil, France
| | - David S Salomon
- Mouse Cancer Genetics Program, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Francis Vacherot
- Inserm, U955, Equipe 7, Créteil, France.,Université Paris-Est, UMR_S955, UPEC, Créteil, France
| |
Collapse
|
33
|
Lefty1 alleviates renal tubulointerstitial injury in mice with unilateral ureteral obstruction. Mol Med Rep 2015; 13:901-8. [DOI: 10.3892/mmr.2015.4631] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 08/27/2015] [Indexed: 11/05/2022] Open
|
34
|
Li FF, Han Y, Shi S, Li X, Zhu XD, Zhou J, Shao QL, Li XQ, Liu SL. Characterization of Transcriptional Repressor Gene MSX1 Variations for Possible Associations with Congenital Heart Diseases. PLoS One 2015; 10:e0142666. [PMID: 26556783 PMCID: PMC4640503 DOI: 10.1371/journal.pone.0142666] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/26/2015] [Indexed: 01/26/2023] Open
Abstract
Background The human heart consists of several cell types with distinct lineage origins. Interactions between these cardiac progenitors are very important for heart formation. The muscle segment homeobox gene family plays a key role in the cell morphogenesis and growth, controlled cellular proliferation, differentiation, and apoptosis, but the relationships between the genetic abnormalities and CHD phenotypes still remain largely unknown. The aim of this work was to evaluate variations in MSX1 and MSX2 for their possible associations with CHD. Methods We sequenced the MSX1 and MSX2 genes for 300 Chinese Han CHD patients and 400 normal controls and identified the variations. The statistical analyses were conducted using Chi-Square Tests as implemented in SPSS (version 19.0). The Hardy-Weinberg equilibrium test of the population was carried out using the online software OEGE. Results Six variations rs4647952, rs2048152, rs4242182, rs61739543, rs111542301 and rs3087539 were identified in the MSX2 gene, but the genetic heterozygosity of those SNPs was very low. In contrast, the genetic heterozygosity of two variations rs3821949 near the 5’UTR and rs12532 within 3’UTR of the MSX1 gene was considerably high. Statistical analyses showed that rs3821949 and rs12532 were associated with the risk of CHD (specifically VSD). Conclusions The SNPs rs3821949 and rs12532 in the MSX1 gene were associated with CHD in Chinese Han populations.
Collapse
Affiliation(s)
- Fei-Feng Li
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Ying Han
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuai Shi
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xia Li
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
| | - Xi-Dong Zhu
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jing Zhou
- Intensive care unit, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qing-Liang Shao
- Department of Neonatalogy, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xue-Qi Li
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
- * E-mail: (S-LL); (X-QL)
| | - Shu-Lin Liu
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Canada
- * E-mail: (S-LL); (X-QL)
| |
Collapse
|
35
|
Li FF, Zhou J, Zhao DD, Yan P, Li X, Han Y, Li XS, Wang GY, Yu KJ, Liu SL. Characterization of SMAD3 Gene Variants for Possible Roles in Ventricular Septal Defects and Other Congenital Heart Diseases. PLoS One 2015; 10:e0131542. [PMID: 26110764 PMCID: PMC4482402 DOI: 10.1371/journal.pone.0131542] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 06/03/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Nodal/TGF signaling pathway has an important effect at early stages of differentiation of human embryonic stem cells in directing them to develop into different embryonic lineages. SMAD3 is a key intracellular messenger regulating factor in the Nodal/TGF signaling pathway, playing important roles in embryonic and, particularly, cardiovascular system development. The aim of this work was to find evidence on whether SMAD3 variations might be associated with ventricular septal defects (VSD) or other congenital heart diseases (CHD). METHODS We sequenced the SMAD3 gene for 372 Chinese Han CHD patients including 176 VSD patients and evaluated SNP rs2289263, which is located before the 5'UTR sequence of the gene. The statistical analyses were conducted using Chi-Square Tests as implemented in SPSS (version 13.0). The Hardy-Weinberg equilibrium test of the population was carried out using the online software OEGE. RESULTS Three heterozygous variants in SMAD3 gene, rs2289263, rs35874463 and rs17228212, were identified. Statistical analyses showed that the rs2289263 variant located before the 5'UTR sequence of SMAD3 gene was associated with the risk of VSD (P value=0.013 <0.05). CONCLUSIONS The SNP rs2289263 in the SMAD3 gene is associated with VSD in Chinese Han populations.
Collapse
Affiliation(s)
- Fei-Feng Li
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
| | - Jing Zhou
- Intensive Care Unit, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dan-Dan Zhao
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
| | - Peng Yan
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xia Li
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
| | - Ying Han
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
| | | | - Gui-Yu Wang
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- * E-mail: (GYW); (KJY); (SLL)
| | - Kai-Jiang Yu
- Intensive Care Unit, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- * E-mail: (GYW); (KJY); (SLL)
| | - Shu-Lin Liu
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Canada
- * E-mail: (GYW); (KJY); (SLL)
| |
Collapse
|
36
|
Poletti V, Delli Carri A, Malagoli Tagliazucchi G, Faedo A, Petiti L, Mazza EMC, Peano C, De Bellis G, Bicciato S, Miccio A, Cattaneo E, Mavilio F. Genome-Wide Definition of Promoter and Enhancer Usage during Neural Induction of Human Embryonic Stem Cells. PLoS One 2015; 10:e0126590. [PMID: 25978676 PMCID: PMC4433211 DOI: 10.1371/journal.pone.0126590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/06/2015] [Indexed: 11/21/2022] Open
Abstract
Genome-wide mapping of transcriptional regulatory elements is an essential tool for understanding the molecular events orchestrating self-renewal, commitment and differentiation of stem cells. We combined high-throughput identification of transcription start sites with genome-wide profiling of histones modifications to map active promoters and enhancers in embryonic stem cells (ESCs) induced to neuroepithelial-like stem cells (NESCs). Our analysis showed that most promoters are active in both cell types while approximately half of the enhancers are cell-specific and account for most of the epigenetic changes occurring during neural induction, and most likely for the modulation of the promoters to generate cell-specific gene expression programs. Interestingly, the majority of the promoters activated or up-regulated during neural induction have a “bivalent” histone modification signature in ESCs, suggesting that developmentally-regulated promoters are already poised for transcription in ESCs, which are apparently pre-committed to neuroectodermal differentiation. Overall, our study provides a collection of differentially used enhancers, promoters, transcription starts sites, protein-coding and non-coding RNAs in human ESCs and ESC-derived NESCs, and a broad, genome-wide description of promoter and enhancer usage and of gene expression programs characterizing the transition from a pluripotent to a neural-restricted cell fate.
Collapse
Affiliation(s)
- Valentina Poletti
- Division of Genetics and Cell Biology, Scientific Institute H. San Raffaele, Milan, Italy
- Genethon, Evry, France
| | | | | | - Andrea Faedo
- Department of Biosciences, University of Milano, Milan, Italy
| | - Luca Petiti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Clelia Peano
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Gianluca De Bellis
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Annarita Miccio
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Imagine Institute, Paris, France
| | - Elena Cattaneo
- Department of Biosciences, University of Milano, Milan, Italy
| | - Fulvio Mavilio
- Genethon, Evry, France
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- * E-mail:
| |
Collapse
|
37
|
LeftyA sensitive cytosolic pH regulation and glycolytic flux in Ishikawa human endometrial cancer cells. Biochem Biophys Res Commun 2015; 460:845-9. [PMID: 25838200 DOI: 10.1016/j.bbrc.2015.03.120] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 03/21/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVE LeftyA, a powerful regulator of stemness, embryonic differentiation, and reprogramming of cancer cells, counteracts cell proliferation and tumor growth. Key properties of tumor cells include enhanced glycolytic flux, which is highly sensitive to cytosolic pH and thus requires export of H(+) and lactate. H(+) extrusion is in part accomplished by Na(+)/H(+) exchangers, such as NHE1. An effect of LeftyA on transport processes has, however, never been reported. The present study thus explored whether LeftyA modifies regulation of cytosolic pH (pHi) in Ishikawa cells, a well differentiated endometrial carcinoma cell model. METHODS NHE1 transcript levels were determined by qRT-PCR, NHE1 protein abundance quantified by Western blotting, pHi estimated utilizing (2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein [BCECF] fluorescence, Na(+)/H(+) exchanger activity from Na(+) dependent realkalinization after an ammonium pulse, and lactate concentration in the supernatant utilizing an enzymatic assay and subsequent colorimetry. RESULTS A 2 h treatment with LeftyA (8 ng/ml) significantly decreased NHE1 transcript levels (by 99.6%), NHE1 protein abundance (by 71%), Na(+)/H(+) exchanger activity (by 55%), pHi (from 7.22 ± 0.02 to 7.05 ± 0.02), and lactate release (by 41%). CONCLUSIONS LeftyA markedly down-regulates NHE1 expression, Na(+)/H(+) exchanger activity, pHi, and lactate release in Ishikawa cells. Those effects presumably contribute to cellular reprogramming and growth inhibition.
Collapse
|
38
|
Kong B, Wang W, Esposito I, Friess H, Michalski CW, Kleeff J. Increased expression of Nodal correlates with reduced patient survival in pancreatic cancer. Pancreatology 2015; 15:156-61. [PMID: 25708930 DOI: 10.1016/j.pan.2015.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 01/12/2015] [Accepted: 02/01/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Nodal (nodal growth differentiation factor) and its inhibitor Lefty (left right determination factor), which are ligands of the TGF (transforming growth factor) β superfamily, are responsible for the determination of left-right asymmetry in vertebrates. Nodal/Lefty signaling has been suggested to play a role in the development of metastatic melanoma and breast cancer. However, it remains unclear whether this pathway is also involved in human pancreatic ductal adenocarcinoma (PDAC). METHODS Pancreatic cancer patient specimens with clinical data (n = 54) were used to investigate the clinical significance of Nodal-Lefty signaling. A set of in vitro assays were carried out in a human pancreatic cancer cell line (Colo-357) to assess the functional relevance of Nodal-Lefty signaling. RESULTS Nodal was absent in the human normal pancreas, while Lefty was present in islet cells. Though Nodal and Lefty expression were found in cancer cells at various expression levels, the cancer-associated tubular complexes were particularly positive for Lefty. Survival analysis revealed that high expression of Nodal correlated with reduced patient survival (median survival 17.8 vs 33.0 months, p = 0.013). Cultured pancreatic cancer cell lines expressed Nodal and Lefty at different levels. In vitro functional assays revealed that treatment with human recombinant Nodal inhibited cell growth and increased invasion of Colo-357 pancreatic cancer cells whereas no effect was found upon treatment with recombinant Lefty. CONCLUSION Nodal-Lefty signaling might be involved in the pathogenesis of PDAC as Nodal expression marks a subtype of PDAC with unfavorable prognosis.
Collapse
Affiliation(s)
- Bo Kong
- Department of Surgery, Technische Universität München, Munich, Germany
| | - Weibin Wang
- Department of Surgery, Technische Universität München, Munich, Germany; Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Irene Esposito
- Institute of Pathology, Technische Universität München, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Technische Universität München, Munich, Germany
| | | | - Jörg Kleeff
- Department of Surgery, Technische Universität München, Munich, Germany.
| |
Collapse
|
39
|
Lefty inhibits glioma growth by suppressing Nodal-activated Smad and ERK1/2 pathways. J Neurol Sci 2014; 347:137-42. [DOI: 10.1016/j.jns.2014.09.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 09/13/2014] [Accepted: 09/15/2014] [Indexed: 01/01/2023]
|
40
|
Li H, Li H, Bai L, Yu H. Lefty inhibits in vitro decidualization by regulating P57 and cyclin D1 expressions. Cell Biochem Funct 2014; 32:657-64. [PMID: 25339094 DOI: 10.1002/cbf.3069] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 09/11/2014] [Accepted: 09/15/2014] [Indexed: 01/20/2023]
Abstract
Endometrial decidualization is highly important for successful construction and maintenance of embryo implantation and pregnancy. Lefty gene at different menstrual cycle phases has different expressions, indicating its regulatory significance. To study the mechanism of Lefty in decidualization, human endometrial stromal cells (hESCs) were cultured and induced with medroxyprogesterone acetate (MPA) and 8-bromoadenosine-cAMP (8-Br-cAMP) in vitro as a research model. Our results showed that Lefty1 overexpression inhibited MPA- and 8-Br-cAMP-induced hESC decidualization and significantly reduced the secretion of prolactin (PRL) and insulin-like growth factor-binding protein 1 (IGFBP-1). With the inhibition of Lefty1 expression, hESC decidualization induced by MPA and 8-Br-cAMP became more remarkable, and the secretions of PRL and IGFBP-1 were higher too. Further tests indicated that during the process of decidualization, P57 expression increased, whereas cyclin D1 expression decreased. Although Lefty1 overexpression did not significantly change the expressions of P57 and cyclin D1, inhibition of Lefty1 expression resulted in more evident changes in P57 and cyclin D1 expressions. Meanwhile, cell cycle examination showed that Lefty1 overexpression reduced the cell cycle arrest at G1/S phase in the in vitro hESC decidualization model. Therefore, Lefty1 could regulate the cell cycle via modulating the expressions of P57 and cyclin D1 and then inhibit the decidualization in vitro.
Collapse
Affiliation(s)
- Hong Li
- Obstetric Department, ShengJing Hospital of China Medical University, Shenyang, China; Obstetric Department, The Maternity Hospital of Dalian, Dalian, China
| | | | | | | |
Collapse
|
41
|
Pourrajab F, Babaei Zarch M, BaghiYazdi M, Hekmatimoghaddam S, Zare-Khormizi MR. MicroRNA-based system in stem cell reprogramming; differentiation/dedifferentiation. Int J Biochem Cell Biol 2014; 55:318-28. [PMID: 25150833 DOI: 10.1016/j.biocel.2014.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 08/09/2014] [Accepted: 08/11/2014] [Indexed: 12/26/2022]
Abstract
Stem cells (SCs) have self-renew ability and give rise to committed progenitors of a single or multiple lineages. Elucidating the genetic circuits that govern SCs to self-renew and to differentiate is essential to understand the roles of SCs and promise of these cells in regenerative medicine. MicroRNAs are widespread agents playing critical roles in regulatory networks of transcriptional expression and have been strongly linked with SCs for simultaneous maintenance of pluripotency properties such as self-renewal. This review aims to provide state-of-the-art presentations on microRNA-dependent molecular mechanisms contribute to the maintenance of pluripotency. Understanding the microRNA signature interactions, in conjunction with cell signaling, is critical for development of improved strategies to reprogram differentiated cells or direct differentiation of pluripotent cells.
Collapse
Affiliation(s)
- Fatemeh Pourrajab
- School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Clinical Biochemistry and Molecular Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | | | - Mohammad BaghiYazdi
- School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | | |
Collapse
|
42
|
The multifaceted role of the embryonic gene Cripto-1 in cancer, stem cells and epithelial-mesenchymal transition. Semin Cancer Biol 2014; 29:51-8. [PMID: 25153355 DOI: 10.1016/j.semcancer.2014.08.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/07/2014] [Indexed: 01/04/2023]
Abstract
Cripto-1 (CR-1)/Teratocarcinoma-derived growth factor1 (TDGF-1) is a cell surface glycosylphosphatidylinositol (GPI)-linked glycoprotein that can function either in cis (autocrine) or in trans (paracrine). The cell membrane cis form is found in lipid rafts and endosomes while the trans acting form lacking the GPI anchor is soluble. As a member of the epidermal growth factor (EGF)/Cripto-1-FRL-1-Cryptic (CFC) family, CR-1 functions as an obligatory co-receptor for the transforming growth factor-β (TGF-β) family members, Nodal and growth and differentiation factors 1 and 3 (GDF1/3) by activating Alk4/Alk7 signaling pathways that involve Smads 2, 3 and 4. In addition, CR-1 can activate non-Smad-dependent signaling elements such as PI3K, Akt and MAPK. Both of these pathways depend upon the 78kDa glucose regulated protein (GRP78). Finally, CR-1 can facilitate signaling through the canonical Wnt/β-catenin and Notch/Cbf-1 pathways by functioning as a chaperone protein for LRP5/6 and Notch, respectively. CR-1 is essential for early embryonic development and maintains embryonic stem cell pluripotentiality. CR-1 performs an essential role in the etiology and progression of several types of human tumors where it is expressed in a population of cancer stem cells (CSCs) and facilitates epithelial-mesenchymal transition (EMT). In this context, CR-1 can significantly enhance tumor cell migration, invasion and angiogenesis. Collectively, these facts suggest that CR-1 may be an attractive target in the diagnosis, prognosis and therapy of several types of human cancer.
Collapse
|
43
|
Deng X, Zhou J, Li FF, Yan P, Zhao EY, Hao L, Yu KJ, Liu SL. Characterization of nodal/TGF-lefty signaling pathway gene variants for possible roles in congenital heart diseases. PLoS One 2014; 9:e104535. [PMID: 25111179 PMCID: PMC4128709 DOI: 10.1371/journal.pone.0104535] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 07/09/2014] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Nodal/TGF-Lefty signaling pathway has important effects at early stages of differentiation of human embryonic stem cells in directing them to differentiate into different embryonic lineages. LEFTY, one of transforming growth factors in the Nodal/TGF-Lefty signaling pathway, plays an important role in the development of heart. The aim of this work was to find evidence on whether Lefty variations are associated with congenital heart diseases (CHD). METHODS We sequenced the Lefty gene for 230 Chinese Han CHD patients and evaluated SNPs rs2295418, rs360057 and g.G169A, which are located within the translated regions of the genes. The statistical analyses were conducted using Chi-Square Tests as implemented in SPSS (version 13.0). The Hardy-Weinberg equilibrium test of the population was carried out using online software OEGE, and multiple-sequence alignments of LEFTY proteins were carried out using the Vector NTI software. RESULTS Two heterozygous variants in Lefty1 gene, g.G169A and g.A1035C, and one heterozygous variant in Lefty2 gene, g.C925A, were identified. Statistical analyses showed that the rs2295418 (g.C925A) variant in Lefty2 gene was obviously associated with the risk of CHD (P value = 0.016<0.05). The genotype frequency of rs360057 (g.A1035C) variant in Lefty1 gene was associated with the risk of CHD (P value = 0.007<0.05), but the allele frequency was not (P value = 0.317>0.05). CONCLUSIONS The SNP rs2295418 in the Lefty2 gene is associated with CHD in Chinese Han populations.
Collapse
Affiliation(s)
- Xia Deng
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
| | - Jing Zhou
- Intensive Care Unit, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fei-Feng Li
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
- * E-mail: (K-JY); (F-FL); (S-LL)
| | - Peng Yan
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Er-Ying Zhao
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
| | - Ling Hao
- Department of Oncology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kai-Jiang Yu
- Intensive Care Unit, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- * E-mail: (K-JY); (F-FL); (S-LL)
| | - Shu-Lin Liu
- Genomics Research Center (one of the State-Province Key Laboratory of Biopharmaceutical Engineering, China), Harbin Medical University, Harbin, China
- Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, Canada
- * E-mail: (K-JY); (F-FL); (S-LL)
| |
Collapse
|
44
|
Alvarez AA, Field M, Bushnev S, Longo MS, Sugaya K. The effects of histone deacetylase inhibitors on glioblastoma-derived stem cells. J Mol Neurosci 2014; 55:7-20. [PMID: 24874578 DOI: 10.1007/s12031-014-0329-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 05/09/2014] [Indexed: 12/13/2022]
Abstract
Glioblastoma multiforme (GBM) is the most malignant brain tumor with limited effective treatment options. Cancer stem cells (CSCs), a subpopulation of cancer cells with stem cell properties found in GBMs, have been shown to be extremely resistant to radiation and chemotherapeutic agents and have the ability to readily reform tumors. Therefore, the development of therapeutic agents targeting CSCs is extremely important. In this study, we isolated glioblastoma-derived stem cells (GDSCs) from GBM tissue removed from patients during surgery and analyzed their gene expression using quantitative real-time PCR and immunocytochemistry. We examined the effects of histone deacetylase inhibitors trichostatin A (TSA) and valproic acid (VPA) on the proliferation and gene expression profiles of GDSCs. The GDSCs expressed significantly higher levels of both neural and embryonic stem cell markers compared to GBM cells expanded in conventional monolayer cultures. Treatment of GDSCs with histone deacetylase inhibitors, TSA and VPA, significantly reduced proliferation rates of the cells and expression of the stem cell markers, indicating differentiation of the cells. Since differentiation into GBM makes them susceptible to the conventional cancer treatments, we posit that use of histone deacetylase inhibitors may increase efficacy of the conventional cancer treatments for eliminating GDSCs.
Collapse
|
45
|
Azpiazu R, Amaral A, Castillo J, Estanyol JM, Guimerà M, Ballescà JL, Balasch J, Oliva R. High-throughput sperm differential proteomics suggests that epigenetic alterations contribute to failed assisted reproduction. Hum Reprod 2014; 29:1225-37. [PMID: 24781426 DOI: 10.1093/humrep/deu073] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
STUDY QUESTION Are there quantitative alterations in the proteome of normozoospermic sperm samples that are able to complete IVF but whose female partner does not achieve pregnancy? SUMMARY ANSWER Normozoospermic sperm samples with different IVF outcomes (pregnancy versus no pregnancy) differed in the levels of at least 66 proteins. WHAT IS KNOWN ALREADY The analysis of the proteome of sperm samples with distinct fertilization capacity using low-throughput proteomic techniques resulted in the detection of a few differential proteins. Current high-throughput mass spectrometry approaches allow the identification and quantification of a substantially higher number of proteins. STUDY DESIGN, SIZE, DURATION This was a case-control study including 31 men with normozoospermic sperm and their partners who underwent IVF with successful fertilization recruited between 2007 and 2008. PARTICIPANTS/MATERIALS, SETTING, METHODS Normozoospermic sperm samples from 15 men whose female partners did not achieve pregnancy after IVF (no pregnancy) and 16 men from couples that did achieve pregnancy after IVF (pregnancy) were included in this study. To perform the differential proteomic experiments, 10 no pregnancy samples and 10 pregnancy samples were separately pooled and subsequently used for tandem mass tags (TMT) protein labelling, sodium dodecyl sulphate-polyacrylamide gel electrophoresis, liquid chromatography tandem mass spectrometry (LC-MS/MS) identification and peak intensity relative protein quantification. Bioinformatic analyses were performed using UniProt Knowledgebase, DAVID and Reactome. Individual samples (n = 5 no pregnancy samples; n = 6 pregnancy samples) and aliquots from the above TMT pools were used for western blotting. MAIN RESULTS AND THE ROLE OF CHANCE By using TMT labelling and LC-MS/MS, we have detected 31 proteins present at lower abundance (ratio no pregnancy/pregnancy < 0.67) and 35 at higher abundance (ratio no pregnancy/pregnancy > 1.5) in the no pregnancy group. Bioinformatic analyses showed that the proteins with differing abundance are involved in chromatin assembly and lipoprotein metabolism (P values < 0.05). In addition, the differential abundance of one of the proteins (SRSF protein kinase 1) was further validated by western blotting using independent samples (P value < 0.01). LIMITATIONS, REASONS FOR CAUTION For individual samples the amount of recovered sperm not used for IVF was low and in most of the cases insufficient for MS analysis, therefore pools of samples had to be used to this end. WIDER IMPLICATIONS OF THE FINDINGS Alterations in the proteins involved in chromatin assembly and metabolism may result in epigenetic errors during spermatogenesis, leading to inaccurate sperm epigenetic signatures, which could ultimately prevent embryonic development. These sperm proteins may thus possibly have clinical relevance. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the Spanish Ministry of Economy and Competitiveness (Ministerio de Economia y Competividad; FEDER BFU 2009-07118 and PI13/00699) and Fundación Salud 2000 SERONO13-015. There are no competing interests to declare.
Collapse
Affiliation(s)
- Rubén Azpiazu
- Human Genetics Research Group, IDIBAPS, Faculty of Medicine, University of Barcelona, Casanova 143, Barcelona 08036, Spain
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Park KS, Cha Y, Kim CH, Ahn HJ, Kim D, Ko S, Kim KH, Chang MY, Ko JH, Noh YS, Han YM, Kim J, Song J, Kim JY, Tesar PJ, Lanza R, Lee KA, Kim KS. Transcription elongation factor Tcea3 regulates the pluripotent differentiation potential of mouse embryonic stem cells via the Lefty1-Nodal-Smad2 pathway. Stem Cells 2013; 31:282-92. [PMID: 23169579 DOI: 10.1002/stem.1284] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 10/25/2012] [Indexed: 01/07/2023]
Abstract
Self-renewal and pluripotency are hallmark properties of pluripotent stem cells, including embryonic stem cells (ESCs) and iPS cells. Previous studies revealed the ESC-specific core transcription circuitry and showed that these core factors (e.g., Oct3/4, Sox2, and Nanog) regulate not only self-renewal but also pluripotent differentiation. However, it remains elusive how these two cell states are regulated and balanced during in vitro replication and differentiation. Here, we report that the transcription elongation factor Tcea3 is highly enriched in mouse ESCs (mESCs) and plays important roles in regulating the differentiation. Strikingly, altering Tcea3 expression in mESCs did not affect self-renewal under nondifferentiating condition; however, upon exposure to differentiating cues, its overexpression impaired in vitro differentiation capacity, and its knockdown biased differentiation toward mesodermal and endodermal fates. Furthermore, we identified Lefty1 as a downstream target of Tcea3 and showed that the Tcea3-Lefty1-Nodal-Smad2 pathway is an innate program critically regulating cell fate choices between self-replication and differentiation commitment. Together, we propose that Tcea3 critically regulates pluripotent differentiation of mESCs as a molecular rheostat of Nodal-Smad2/3 signaling.
Collapse
Affiliation(s)
- Kyung-Soon Park
- Department of Biomedical Science, College of Life Science, CHA University, Seoul, Korea.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Maraghechi P, Hiripi L, Tóth G, Bontovics B, Bősze Z, Gócza E. Discovery of pluripotency-associated microRNAs in rabbit preimplantation embryos and embryonic stem-like cells. Reproduction 2013; 145:421-37. [DOI: 10.1530/rep-12-0259] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that regulate multiple biological processes. Increasing experimental evidence implies an important regulatory role of miRNAs during embryonic development and in embryonic stem (ES) cell biology. In the current study, we have described and analyzed the expression profile of pluripotency-associated miRNAs in rabbit embryos and ES-like cells. The rabbit specific ocu-miR-302 and ocu-miR-290 clusters, and three homologs of the human C19MC cluster (ocu-miR-512, ocu-miR-520e, and ocu-miR-498) were identified in rabbit preimplantation embryos and ES-like cells. The ocu-miR-302 cluster was highly similar to its human homolog, while ocu-miR-290 revealed a low level of evolutionary conservation with its mouse homologous cluster. The expression of the ocu-miR-302 cluster began at the 3.5 days post-coitum early blastocyst stage and they stayed highly expressed in rabbit ES-like cells. In contrast, a high expression level of the ocu-miR-290 cluster was detected during preimplantation embryonic development, but a low level of expression was found in rabbit ES-like cells. Differential expression of the ocu-miR-302 cluster and ocu-miR-512 miRNA was detected in rabbit trophoblast and embryoblast. We also found that Lefty has two potential target sites in its 3′UTR for ocu-miR-302a and its expression level increased upon ocu-miR-302a inhibition. We suggest that the expression of the ocu-miR-302 cluster is characteristic of the rabbit ES-like cell, while the ocu-miR-290 cluster may play a crucial role during early embryonic development. This study presents the first identification, to our knowledge, of pluripotency-associated miRNAs in rabbit preimplantation embryos and ES-like cells, which can open up new avenues to investigate the regulatory function of ocu-miRNAs in embryonic development and stem cell biology.
Collapse
|
48
|
Saito A, Ochiai H, Okada S, Miyata N, Azuma T. Suppression of Lefty expression in induced pluripotent cancer cells. FASEB J 2013; 27:2165-74. [PMID: 23407711 DOI: 10.1096/fj.12-221432] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cancer and stem cells share the ability to silence tumor suppressors. We focused on Lefty, which encodes one of the most abundant tumor suppressors in embryonic stem (ES) cells and is not expressed in somatic cancer cells. We found that transforming growth factor β (TGF-β) induced demethylation of the Lefty B cytosine-phosphate-guanine (CpG) island and increased Lefty expression (10-200 times) in human pancreatic cancer cells and human liver cancer cells (PLC/PRF/5 and HLF). Expression of Cripto, another important factor in Nodal-Lefty signaling, was not increased after adding TGF-β. We generated reprogrammed cancer cells that revealed high expression of immature marker proteins, high proliferation, and the potential to express morphological patterns of ectoderm, mesoderm, and endoderm, suggesting that these cells may have cancer stem cell-like phenotypes. We investigated Lefty and found that reprogrammed human liver cancer cells (induced pluripotent cancer cells) displayed a much lower ability to express Lefty, although less Lefty B CpG methylation was also observed. We also found that a MEK inhibitor dramatically enhanced Lefty expression in human pancreatic cancers with mutated ras, whereas Lefty B CpG methylation was not decreased. These observations indicate that despite the demethylation of DNA strands in promoter regions of pluripotency-associated genes, including Lefty gene, Lefty expression was not induced well in reprogrammed cells. Of note was the fact that Lefty is abundantly expressed in human ES cells but not in induced pluripotent stem (iPS) cells. We thus think that reprogrammed cancer cells share the mechanism for expression of Lefty with iPS cells. This shared mechanism may contribute to the cancerous transformation of iPS cells.
Collapse
Affiliation(s)
- Akiko Saito
- Department of Biochemistry, Tokyo Dental College, Chiba, Japan
| | | | | | | | | |
Collapse
|
49
|
Park CB, Dufort D. NODAL signaling components regulate essential events in the establishment of pregnancy. Reproduction 2013; 145:R55-64. [DOI: 10.1530/rep-12-0103] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Successful mammalian reproduction is dependent on a receptive and nurturing uterine environment. In order to establish pregnancy in humans, the uterus must i) be adequately prepared to receive the blastocyst, ii) engage in a coordinated molecular dialog with the embryo to facilitate implantation, and iii) undergo endometrial decidualization. Although numerous factors have been implicated in these essential processes, the precise network of molecular interactions that govern receptivity, embryo implantation, and decidualization remain unclear. NODAL, a morphogen in the transforming growth factor β superfamily, is well known for its critical functions during embryogenesis; however, recent studies have demonstrated an emerging role for NODAL signaling during early mammalian reproduction. Here, we review the established data and a recent wave of new studies implicating NODAL signaling components in uterine cycling, embryo implantation, and endometrial decidualization in humans and mice.
Collapse
|
50
|
Quail DF, Siegers GM, Jewer M, Postovit LM. Nodal signalling in embryogenesis and tumourigenesis. Int J Biochem Cell Biol 2013; 45:885-98. [PMID: 23291354 DOI: 10.1016/j.biocel.2012.12.021] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 12/19/2012] [Accepted: 12/24/2012] [Indexed: 12/22/2022]
Abstract
With few exceptions, most cells in adult organisms have lost the expression of stem cell-associated proteins and are instead characterized by tissue-specific gene expression and function. This cell fate specification is dictated spatially and temporally during embryogenesis. It has become increasingly apparent that the elegant and complicated process of cell specification is "undone" in cancer. This may be because cancer cells respond to their microenvironment and mutations by acquiring a more permissive, plastic epigenome, or because cancer cells arise from mutated stem cells. Regardless, these advanced cancer cells must use stem cell-associated proteins to sustain their phenotype. One such protein is Nodal, an embryonic morphogen belonging to the transforming growth factor-β (TGF-β) superfamily. First described in early developmental models, Nodal orchestrates embryogenesis by regulating a myriad of processes, including mesendoderm induction, left-right asymmetry and embryo implantation. Nodal is relatively restricted to embryonic and reproductive cell types and is thus absent from most normal adult tissues. However, recent studies focusing on a variety of malignancies have demonstrated that Nodal expression re-emerges during cancer progression. Moreover, in almost every cancer studied thus far, the acquisition of Nodal expression is associated with increased tumourigenesis, invasion and metastasis. As the list of cancers that express Nodal grows, it is essential that the scientific and medical communities fully understand how this morphogen is regulated in both normal and neoplastic conditions. Herein, we review the literature relating to normal and pathological Nodal signalling. In particular, we emphasize the role that this secreted protein plays during morphogenic events and how it signals to support stem cell maintenance and tumour progression.
Collapse
Affiliation(s)
- Daniela F Quail
- Department of Anatomy and Cell Biology, University of Western Ontario and Robarts Research Institute, London, ON, Canada
| | | | | | | |
Collapse
|