1
|
Retention of ERK in the cytoplasm mediates the pluripotency of embryonic stem cells. Stem Cell Reports 2022; 18:305-318. [PMID: 36563690 PMCID: PMC9860118 DOI: 10.1016/j.stemcr.2022.11.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 11/15/2022] [Accepted: 11/19/2022] [Indexed: 12/24/2022] Open
Abstract
The dynamic subcellular localization of ERK1/2 plays an important role in regulating cell fate. Differentiation of mouse embryonic stem cells (mESCs) involves inductive stimulation of ERK1/2, and therefore, inhibitors of the ERK cascade are used to maintain pluripotency. Interestingly, we found that in pluripotent mESCs, ERK1/2 do not translocate to the nucleus either before or after stimulation. This inhibition of nuclear translocation may be dependent on a lack of stimulated ERK1/2 interaction with importin7 rather than a lack of ERK1/2 phosphorylation activating translocation. At late stages of naive-to-primed transition, the action of the translocating machinery is restored, leading to elevation in ERK1/2-importin7 interaction and their nuclear translocation. Importantly, forcing ERK2 into the naive cells' nuclei accelerates their early differentiation, while prevention of the translocation restores stem cells' pluripotency. These results indicate that prevention of nuclear ERK1/2 translocation serves as a safety mechanism for keeping pluripotency of mESCs.
Collapse
|
2
|
Zhang Y, Zhang H, Xiao Z, Yuan G, Yang G. IPO7 Promotes Odontoblastic Differentiation and Inhibits Osteoblastic Differentiation Through Regulation of RUNX2 Expression and Translocation. Stem Cells 2022; 40:1020-1030. [PMID: 35922041 DOI: 10.1093/stmcls/sxac055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022]
Abstract
RUNX2, an important transcriptional factor for both odontoblastic and osteoblastic differentiation, is upregulated during osteoblastic differentiation, but downregulated during late odontoblastic differentiation. However, the specific mechanism of the different RUNX2 expression in bone and dentin remains largely unknown. Importin 7 (IPO7), a member of the karyopherin β-superfamily, mediates nucleocytoplasmic transport of proteins. In this study, we found that IPO7 was increasingly expressed from pre-odontoblasts to mature odontoblasts. IPO7 expression was increased with odontoblastic differentiation of mouse dental papilla cells (mDPCs) and knockdown of IPO7-inhibited cell differentiation. While in MC3T3-E1 cells, IPO7 was decreased during osteoblastic differentiation and knockdown of IPO7-promoted cell differentiation. In mPDCs, IPO7 was able to bind with some odontoblastic transcription factors, and imported them into the nucleus, but not with RUNX2. Furthermore, IPO7 inhibited the total RUNX2 expression by promoting HDAC6 nuclear localization during odontoblastic differentiation. However, in MC3T3-E1 cells, IPO7 inhibited the nuclear distribution of RUNX2 but did not affect the total protein level of RUNX2. In conclusion, we found that IPO7 promotes odontoblastic differentiation and inhibits osteoblastic differentiation through regulating RUNX2 expression and translocation differently.
Collapse
Affiliation(s)
- Yue Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, People's Republic of China
| | - Hao Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, People's Republic of China
| | - Ziqiu Xiao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, People's Republic of China
| | - Guohua Yuan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, People's Republic of China
| | - Guobin Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
3
|
Mandic R, Marquardt A, Terhorst P, Ali U, Nowak-Rossmann A, Cai C, Rodepeter FR, Stiewe T, Wezorke B, Wanzel M, Neff A, Stuck BA, Bette M. The importin beta superfamily member RanBP17 exhibits a role in cell proliferation and is associated with improved survival of patients with HPV+ HNSCC. BMC Cancer 2022; 22:785. [PMID: 35850701 PMCID: PMC9290296 DOI: 10.1186/s12885-022-09854-0] [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: 11/12/2021] [Accepted: 06/29/2022] [Indexed: 11/10/2022] Open
Abstract
Background More than twenty years after its discovery, the role of the importin beta superfamily member Ran GTP-binding protein (RanBP) 17 is still ill defined. Previously, we observed notable RanBP17 RNA expression levels in head and neck squamous cell carcinoma (HNSCC) cell lines with disruptive TP53 mutations. Methods We deployed HNSCC cell lines as well as cell lines from other tumor entities such as HCT116, MDA-MB-231 and H460, which were derived from colon, breast and lung cancers respectively. RNAi was used to evaluate the effect of RanBP17 on cell proliferation. FACS analysis was used for cell sorting according to their respective cell cycle phase and for BrdU assays. Immunocytochemistry was deployed for colocalization studies of RanBP17 with Nucleolin and SC35 (nuclear speckles) domains. TCGA analysis was performed for prognostic assessment and correlation analysis of RanBP17 in HNSCC patients. Results RNAi knockdown of RanBP17, significantly reduced cell proliferation in HNSCC cell lines. This effect was also seen in the HNSCC unrelated cell lines HCT116 and MDA-MB-231. Similarly, inhibiting cell proliferation with cisplatin reduced RanBP17 in keratinocytes but lead to induction in tumor cell lines. A similar observation was made in tumor cell lines after treatment with the EGFR kinase inhibitor AG1478. In addition to previous reports, showing colocalization of RanBP17 with SC35 domains, we observed colocalization of RanBP17 to nuclear bodies that are distinct from nucleoli and SC35 domains. Interestingly, for HPV positive but not HPV negative HNSCC, TCGA data base analysis revealed a strong positive correlation of RanBP17 RNA with patient survival and CDKN2A. Conclusions Our data point to a role of RanBP17 in proliferation of HNSCC and other epithelial cells. Furthermore, RanBP17 could potentially serve as a novel prognostic marker for HNSCC patients. However, we noted a major discrepancy between RanBP17 RNA and protein expression levels with the used antibodies. These observations could be explained by the presence of additional RanBP17 splice isoforms and more so of non-coding circular RanBP17 RNA species. These aspects need to be addressed in more detail by future studies. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09854-0.
Collapse
Affiliation(s)
- Robert Mandic
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Giessen and Marburg, Campus Marburg, Philipps-Universität Marburg, 3. BA, +3/08070, Marburg, Germany.
| | - André Marquardt
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany.,Institute of Pathology, University of Würzburg, Würzburg, Germany.,Bavarian Cancer Research Center (BZKF), Würzburg, Germany
| | - Philip Terhorst
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Giessen and Marburg, Campus Marburg, Philipps-Universität Marburg, 3. BA, +3/08070, Marburg, Germany
| | - Uzma Ali
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Giessen and Marburg, Campus Marburg, Philipps-Universität Marburg, 3. BA, +3/08070, Marburg, Germany.,Institute for Pharmaceutical Technology & Biopharmacy, Philipps-Universität Marburg, Marburg, Germany
| | - Annette Nowak-Rossmann
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Giessen and Marburg, Campus Marburg, Philipps-Universität Marburg, 3. BA, +3/08070, Marburg, Germany.,Department of Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps-Universität Marburg, Marburg, Germany
| | - Chengzhong Cai
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Giessen and Marburg, Campus Marburg, Philipps-Universität Marburg, 3. BA, +3/08070, Marburg, Germany
| | - Fiona R Rodepeter
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Giessen and Marburg, Campus Marburg, Philipps-Universität Marburg, 3. BA, +3/08070, Marburg, Germany.,Institute of Pathology, University Hospital Giessen and Marburg, Campus Marburg, Marburg, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-Universität Marburg, Marburg, Germany
| | - Bernadette Wezorke
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-Universität Marburg, Marburg, Germany
| | - Michael Wanzel
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-Universität Marburg, Marburg, Germany
| | - Andreas Neff
- Department of Oro- and Maxillofacial Surgery, University Hospital Giessen and Marburg, Campus Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Boris A Stuck
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Giessen and Marburg, Campus Marburg, Philipps-Universität Marburg, 3. BA, +3/08070, Marburg, Germany
| | - Michael Bette
- Department of Molecular Neuroscience, Institute of Anatomy and Cell Biology, Philipps-Universität Marburg, Marburg, Germany
| |
Collapse
|
4
|
Nuclear Transporter IPO13 Is Central to Efficient Neuronal Differentiation. Cells 2022; 11:cells11121904. [PMID: 35741036 PMCID: PMC9221400 DOI: 10.3390/cells11121904] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 05/28/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022] Open
Abstract
Molecular transport between the nucleus and cytoplasm of the cell is mediated by the importin superfamily of transport receptors, of which the bidirectional transporter Importin 13 (IPO13) is a unique member, with a critical role in early embryonic development through nuclear transport of key regulators, such as transcription factors Pax6, Pax3, and ARX. Here, we examined the role of IPO13 in neuronal differentiation for the first time, using a mouse embryonic stem cell (ESC) model and a monolayer-based differentiation protocol to compare IPO13−/− to wild type ESCs. Although IPO13−/− ESCs differentiated into neural progenitor cells, as indicated by the expression of dorsal forebrain progenitor markers, reduced expression of progenitor markers Pax6 and Nestin compared to IPO13−/− was evident, concomitant with reduced nuclear localisation/transcriptional function of IPO13 import cargo Pax6. Differentiation of IPO13−/− cells into neurons appeared to be strongly impaired, as evidenced by altered morphology, reduced expression of key neuronal markers, and altered response to the neurotransmitter glutamate. Our findings establish that IPO13 has a key role in ESC neuronal differentiation, in part through the nuclear transport of Pax6.
Collapse
|
5
|
She Y, Zhang Y, Xiao Z, Yuan G, Yang G. The regulation of Msx1 by BMP4/pSmad1/5 signaling is mediated by importin7 in dental mesenchymal cells. Cells Dev 2022; 169:203763. [PMID: 34995814 DOI: 10.1016/j.cdev.2021.203763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 02/08/2023]
Abstract
Msx1 is essential for the maintenance of the odontogenic fate of dental mesenchymal cells, and is regulated by BMP/Smad1/5 signaling in a Smad4-independent manner. However, the exact co-factors that assist pSmad1/5 entering the nucleus to regulate Msx1 in dental mesenchymal cells are still unknown. Importin7 (IPO7) is one of the important members of importin β-superfamily, which is mainly responsible for nucleocytoplasmic shuttling of RNAs and proteins, including transcription factors. This study aims to investigate whether IPO7 participates in the nuclear translocation of pSmad1/5 activated by BMP4 to regulate Msx1 expression in mouse dental mesenchymal cells. In the current study, we found that IPO7 was strongly expressed in the mouse dental mesenchymal cells at postnatal day 1 (PN1) both in vitro and in vivo. With BMP4 stimulation, IPO7 showed a translocation from the cytoplasm to the nucleus. Knockdown of IPO7 with siRNA inhibited the nuclear accumulation of pSmad1/5 in response to BMP4 stimulation. Furthermore, the co-immunoprecipitation assay showed pSmad1/5 was a nuclear import cargo of IPO7. Next, knockdown of IPO7 abolished the upregulation of Msx1 induced by BMP4, while overexpression of Smad1 was able to rescue the Msx1 expression. Finally, ChIP and Re-ChIP assay showed IPO7 facilitated the recruitment of pSmad1/5 to the Msx1 promoter. Taken together, our data demonstrated that the regulation of Msx1 by BMP4/pSmad1/5 signaling is mediated by importin7 in mouse dental mesenchymal cells.
Collapse
Affiliation(s)
- Yawei She
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yue Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ziqiu Xiao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guohua Yuan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guobin Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
6
|
Lasorsa VA, Montella A, Cantalupo S, Tirelli M, de Torres C, Aveic S, Tonini GP, Iolascon A, Capasso M. Somatic mutations enriched in cis-regulatory elements affect genes involved in embryonic development and immune system response in neuroblastoma. Cancer Res 2022; 82:1193-1207. [PMID: 35101866 DOI: 10.1158/0008-5472.can-20-3788] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/04/2021] [Accepted: 01/27/2022] [Indexed: 11/16/2022]
Abstract
Noncoding cis-regulatory variants have gained interest as cancer drivers, yet progress in understanding their significance is hindered by the numerous challenges and limitations of variant prioritization. To overcome these limitations, we focused on active cis-regulatory elements (aCRE) in order to design a customized panel for the deep sequencing of 56 neuroblastoma tumor and normal DNA sample pairs. In order to search for driver mutations, aCREs were defined by reanalysis of H3K27ac ChiP-seq peaks in 25 neuroblastoma cell lines. These regulatory genomic regions were tested for an excess of somatic mutations and assessed for statistical significance using a global approach that accounted for chromatin accessibility and replication timing. Additional validation was provided by whole genome sequence analysis of 151 neuroblastomas. Analysis of Hi-C data determined the presence of candidate target genes interacting with mutated regions. An excess of somatic mutations in aCREs of diverse genes were identified, including IPO7, HAND2, and ARID3A. CRISPR-Cas9 editing was utilized to assess the functional consequences of mutations in the IPO7 aCRE. Patients with noncoding mutations in aCREs showed inferior overall and event-free survival independent of age at diagnosis, stage, risk stratification, and MYCN status. Expression of aCRE-interacting genes correlated strongly with negative prognostic markers and low survival rates. Moreover, a convergence between the biological functions of aCRE target genes and transcription factors with mutated binding motifs was associated with embryonic development and immune system response. Overall, this strategy enabled the identification of somatic mutations in regulatory elements that collectively promote neuroblastoma tumorigenesis.
Collapse
Affiliation(s)
- Vito Alessandro Lasorsa
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II
| | - Annalaura Montella
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II di Napoli, CEINGE Biotecnologie Avanzate
| | | | | | - Carmen de Torres
- Developmental Tumor Biology Laboratory and Department of Oncology, Hospital Sant Joan de Déu Barcelona
| | - Sanja Aveic
- Neuroblastoma Laboratory, Fondazione Istituto di Ricerca Pediatrica Citta della Speranza
| | | | - Achille Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II
| | - Mario Capasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II
| |
Collapse
|
7
|
Zhang Y, Zhang H, Yuan G, Yang G. Effects of transforming growth factor-β1 on odontoblastic differentiation in dental papilla cells is determined by IPO7 expression level. Biochem Biophys Res Commun 2021; 545:105-111. [PMID: 33548622 DOI: 10.1016/j.bbrc.2021.01.076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 01/20/2021] [Indexed: 12/25/2022]
Abstract
Transforming growth factor β1 (TGF-β1) is one of the broad-spectrum growth-promoting factors that participate in tooth development. The influence of TGF-β1 on the odontoblastic differentiation is still controvercy. Mouse primary dental papilla cells (mDPCs) as well as an immortalized mouse dental papilla cell line (mDPC6Ts) were treated with exogenous TGF-β1 during odontoblastic differentiation. RT-qPCR, Western blot, alizarin red staining and ALP staining were carried out to investigate the influence of TGF-β1 on odontoblastic differentiation. IPO7, important for SMAD complex translocation was also detected in mDPCs and mDPC6Ts in response to TGF-β1. After silencing IPO7 by transfection, the translocation process of P-SMAD2 was investigated by nuclear and cytoplasmic extraction as well as co-immunoprecipitation assay. The odontogenic markers, mineralization and IPO7 expression were significantly up-regulated in TGF-β1-treated mDPCs while down-regulated in mDPC6Ts. The total level of P-SMAD2 was not influenced by IPO7 in mDPCs, however, IPO7 could bind to P-SMAD2 and affect the nuclear-cytoplasm-shuttling of P-SMAD2. Our data demonstrated that TGF-β1 plays opposite roles in odontoblast differentiation in mDPCs and immortalized mouse dental papilla cell line (mDPC6Ts), which is determined by IPO7.
Collapse
Affiliation(s)
- Yue Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, China.
| | - Hao Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, China.
| | - Guohua Yuan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, China.
| | - Guobin Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, China.
| |
Collapse
|
8
|
Khan AU, Qu R, Ouyang J, Dai J. Role of Nucleoporins and Transport Receptors in Cell Differentiation. Front Physiol 2020; 11:239. [PMID: 32308628 PMCID: PMC7145948 DOI: 10.3389/fphys.2020.00239] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 03/02/2020] [Indexed: 12/20/2022] Open
Abstract
Bidirectional molecular movements between the nucleus and cytoplasm take place through nuclear pore complexes (NPCs) embedded in the nuclear membrane. These macromolecular structures are composed of several nucleoporins, which form seven different subcomplexes based on their biochemical affinity. These nucleoporins are integral components of the complex, not only allowing passive transport but also interacting with importin, exportin, and other molecules that are required for transport of protein in various cellular processes. Transport of different proteins is carried out either dependently or independently on transport receptors. As well as facilitating nucleocytoplasmic transport, nucleoporins also play an important role in cell differentiation, possibly by their direct gene interaction. This review will cover the general role of nucleoporins (whether its dependent or independent) and nucleocytoplasmic transport receptors in cell differentiation.
Collapse
|
9
|
Hsu CY, Salazar MG, Miller S, Meyers C, Ding C, Hardy W, Péault B, James AW. Comparison of Human Tissue Microarray to Human Pericyte Transcriptome Yields Novel Perivascular Cell Markers. Stem Cells Dev 2019; 28:1214-1223. [PMID: 31264500 DOI: 10.1089/scd.2019.0106] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Human perivascular progenitor cells, including pericytes, are well-described multipotent mesenchymal cells giving rise to mesenchymal stem cells in culture. Despite the unique location of pericytes, specific antigens to distinguish human pericytes from other cell types are few. Here, we employed a human tissue microarray (Human Protein Atlas) to identify proteins that are strongly and specifically expressed in a pericytic location within human adipose tissue. Next, these results were cross-referenced with RNA sequencing data from human adipose tissue pericytes, as defined as a fluorescence activated cell sorting (FACS) purified CD146+CD34-CD31-CD45- cell population. Results showed that from 105,532 core biopsies of soft tissue, 229 proteins showed strong and specific perivascular immunoreactivity, the majority of which (155) were present in the tunica intima. Next, cross-referencing with the transcriptome of FACS-derived CD146+ pericytes yielded 25 consistently expressed genes/proteins, including 18 novel antigens. A majority of these transcripts showed maintained expression after culture propagation (56% of genes). Interestingly, many novel antigens within pericytes are regulators of osteogenic differentiation. In sum, our study demonstrates the existence of novel pericyte markers, some of which are conserved in culture that may be useful for future efforts to typify, isolate, and characterize human pericytes.
Collapse
Affiliation(s)
- Ching Yun Hsu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Mario Gomez Salazar
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom.,MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Sarah Miller
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Carolyn Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Catherine Ding
- Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Winters Hardy
- Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Bruno Péault
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom.,MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom.,Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland.,Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
10
|
Zhou D, Alver BM, Li S, Hlady RA, Thompson JJ, Schroeder MA, Lee JH, Qiu J, Schwartz PH, Sarkaria JN, Robertson KD. Distinctive epigenomes characterize glioma stem cells and their response to differentiation cues. Genome Biol 2018; 19:43. [PMID: 29587824 PMCID: PMC5872397 DOI: 10.1186/s13059-018-1420-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/12/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Glioma stem cells (GSCs) are a subpopulation of stem-like cells that contribute to glioblastoma (GBM) aggressiveness, recurrence, and resistance to radiation and chemotherapy. Therapeutically targeting the GSC population may improve patient survival, but unique vulnerabilities need to be identified. RESULTS We isolate GSCs from well-characterized GBM patient-derived xenografts (PDX), characterize their stemness properties using immunofluorescence staining, profile their epigenome including 5mC, 5hmC, 5fC/5caC, and two enhancer marks, and define their transcriptome. Fetal brain-derived neural stem/progenitor cells are used as a comparison to define potential unique and common molecular features between these different brain-derived cells with stem properties. Our integrative study reveals that abnormal expression of ten-eleven-translocation (TET) family members correlates with global levels of 5mC and 5fC/5caC and may be responsible for the distinct levels of these marks between glioma and neural stem cells. Heterogenous transcriptome and epigenome signatures among GSCs converge on several genes and pathways, including DNA damage response and cell proliferation, which are highly correlated with TET expression. Distinct enhancer landscapes are also strongly associated with differential gene regulation between glioma and neural stem cells; they exhibit unique co-localization patterns with DNA epigenetic mark switching events. Upon differentiation, glioma and neural stem cells exhibit distinct responses with regard to TET expression and DNA mark changes in the genome and GSCs fail to properly remodel their epigenome. CONCLUSIONS Our integrative epigenomic and transcriptomic characterization reveals fundamentally distinct yet potentially targetable biologic features of GSCs that result from their distinct epigenomic landscapes.
Collapse
Affiliation(s)
- Dan Zhou
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Bonnie M Alver
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Shuang Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Ryan A Hlady
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Joyce J Thompson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Mark A Schroeder
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Jeong-Heon Lee
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.,Epigenomics Translational Program, Mayo Clinic, Rochester, MN, USA
| | - Jingxin Qiu
- Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Philip H Schwartz
- National Human Neural Stem Cell Resource, Children's Hospital of Orange County Research Institute, Orange, CA, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Keith D Robertson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA. .,Epigenomics Translational Program, Mayo Clinic, Rochester, MN, USA. .,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA. .,Mayo Clinic Cancer Center, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
11
|
Reprint of: Importins in the maintenance and lineage commitment of ES cells. Neurochem Int 2017; 106:14-23. [PMID: 28550879 DOI: 10.1016/j.neuint.2017.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 01/25/2017] [Accepted: 01/30/2017] [Indexed: 11/23/2022]
Abstract
The nucleus of a eukaryotic cell is separated from the cytoplasm by a nuclear envelope, and nuclear pores within the envelope facilitate nucleocytoplasmic transport and the exchange of information. Gene regulation is a key component of biological activity regulation in the cell. Transcription factors control the expression levels of various genes that are necessary for the maintenance or conversion of cellular states during animal development. Because transcription factor activities determine the extent of transcription of target genes, the number of active transcription factors must be tightly regulated. In this regard, the nuclear translocation of a transcription factor is an important determinant of its activity. Therefore, it is becoming clear that the nucleocytoplasmic transport machinery is involved in cell differentiation and organism development. This review examines the regulation of transcription factors by the nucleocytoplasmic transport machinery in ES cells.
Collapse
|
12
|
Competitive regulation of IPO4 transcription by ELK1 and GABP. Gene 2017; 613:30-38. [DOI: 10.1016/j.gene.2017.02.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/25/2017] [Accepted: 02/24/2017] [Indexed: 11/19/2022]
|
13
|
Importins in the maintenance and lineage commitment of ES cells. Neurochem Int 2017; 105:32-41. [PMID: 28163061 DOI: 10.1016/j.neuint.2017.01.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 01/25/2017] [Accepted: 01/30/2017] [Indexed: 11/23/2022]
Abstract
The nucleus of a eukaryotic cell is separated from the cytoplasm by a nuclear envelope, and nuclear pores within the envelope facilitate nucleocytoplasmic transport and the exchange of information. Gene regulation is a key component of biological activity regulation in the cell. Transcription factors control the expression levels of various genes that are necessary for the maintenance or conversion of cellular states during animal development. Because transcription factor activities determine the extent of transcription of target genes, the number of active transcription factors must be tightly regulated. In this regard, the nuclear translocation of a transcription factor is an important determinant of its activity. Therefore, it is becoming clear that the nucleocytoplasmic transport machinery is involved in cell differentiation and organism development. This review examines the regulation of transcription factors by the nucleocytoplasmic transport machinery in ES cells.
Collapse
|
14
|
Taleahmad S, Mirzaei M, Parker LM, Hassani SN, Mollamohammadi S, Sharifi-Zarchi A, Haynes PA, Baharvand H, Salekdeh GH. Proteome Analysis of Ground State Pluripotency. Sci Rep 2015; 5:17985. [PMID: 26671762 PMCID: PMC4680864 DOI: 10.1038/srep17985] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 11/09/2015] [Indexed: 02/07/2023] Open
Abstract
The differentiation potential of pluripotent embryonic stem cells (ESCs) can be manipulated via serum and medium conditions for direct cellular development or to maintain a naïve ground state. The self-renewal state of ESCs can thus be induced by adding inhibitors of mitogen activated protein kinase (MAPK) and glycogen synthase kinase-3 (Gsk3), known as 2 inhibitors (2i) treatment. We have used a shotgun proteomics approach to investigate differences in protein expressions between 2i- and serum-grown mESCs. The results indicated that 164 proteins were significantly upregulated and 107 proteins downregulated in 2i-grown cells compared to serum. Protein pathways in 2i-grown cells with the highest enrichment were associated with glycolysis and gluconeogenesis. Protein pathways related to organ development were downregulated in 2i-grown cells. In serum-grown ESCs, protein pathways involved in integrin and focal adhesion, and signaling proteins involved in the actin cytoskeleton regulation were enriched. We observed a number of nuclear proteins which were mostly involved in self-renewal maintenance and were expressed at higher levels in 2i compared to serum - Dnmt1, Map2k1, Parp1, Xpo4, Eif3g, Smarca4/Brg1 and Smarcc1/Baf155. Collectively, the results provided an insight into the key protein pathways used by ESCs in the ground state or metastable conditions through 2i or serum culture medium, respectively.
Collapse
Affiliation(s)
- Sara Taleahmad
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mehdi Mirzaei
- Department of Chemistry and Biomolecular sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Lindsay M Parker
- Department of Chemistry and Biomolecular sciences, Macquarie University, Sydney, NSW, 2109, Australia.,ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), Macquarie University, NSW, 2109, Sydney, Australia
| | - Seyedeh-Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sepideh Mollamohammadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ali Sharifi-Zarchi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Paul A Haynes
- Department of Chemistry and Biomolecular sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Hossein Baharvand
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), Macquarie University, NSW, 2109, Sydney, Australia.,Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Ghasem Hosseini Salekdeh
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Systems Biology, Agricultural Biotechnology Research Institute of Iran, Karaj, Iran
| |
Collapse
|