1
|
Okubo C, Nakamura M, Sato M, Shichino Y, Mito M, Takashima Y, Iwasaki S, Takahashi K. EIF3D safeguards the homeostasis of key signaling pathways in human primed pluripotency. SCIENCE ADVANCES 2025; 11:eadq5484. [PMID: 40203091 PMCID: PMC11980838 DOI: 10.1126/sciadv.adq5484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Although pluripotent stem cell (PSC) properties, such as differentiation and infinite proliferation, have been well documented within the frameworks of transcription factor networks, epigenomes, and signal transduction, they remain unclear and fragmented. Directing attention toward translational regulation as a bridge between these events can yield additional insights into previously unexplained mechanisms. Our functional CRISPR interference screen-based approach revealed that EIF3D, a translation initiation factor, is crucial for maintaining primed pluripotency. Loss of EIF3D disrupted the balance of pluripotency-associated signaling pathways, thereby compromising primed pluripotency. Moreover, EIF3D ensured robust proliferation by controlling the translation of various p53 regulators, which maintain low p53 activity in the undifferentiated state. In this way, EIF3D-mediated translation contributes to tuning the homeostasis of the primed pluripotency networks, ensuring the maintenance of an undifferentiated state with high proliferative potential. This study provides further insights into the translation network in maintaining pluripotency.
Collapse
Affiliation(s)
- Chikako Okubo
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Michiko Nakamura
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Masae Sato
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Yuichi Shichino
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Saitama, 351-0198, Japan
| | - Mari Mito
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Saitama, 351-0198, Japan
| | - Yasuhiro Takashima
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Shintaro Iwasaki
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Saitama, 351-0198, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, 277-8561, Japan
| | - Kazutoshi Takahashi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| |
Collapse
|
2
|
Xiang J, Wang H, Shi B, Li J, Liu D, Wang K, Wang Z, Min Q, Zhao C, Pei D. Pig blastocyst-like structure models from embryonic stem cells. Cell Discov 2024; 10:72. [PMID: 38956027 PMCID: PMC11219778 DOI: 10.1038/s41421-024-00693-w] [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: 09/06/2023] [Accepted: 05/24/2024] [Indexed: 07/04/2024] Open
Abstract
Pluripotent stem cells have the potential to generate embryo models that can recapitulate developmental processes in vitro. Large animals such as pigs may also benefit from stem-cell-based embryo models for improving breeding. Here, we report the generation of blastoids from porcine embryonic stem cells (pESCs). We first develop a culture medium 4FIXY to derive pESCs. We develop a 3D two-step differentiation strategy to generate porcine blastoids from the pESCs. The resulting blastoids exhibit similar morphology, size, cell lineage composition, and single-cell transcriptome characteristics to blastocysts. These porcine blastoids survive and expand for more than two weeks in vitro under two different culture conditions. Large animal blastoids such as those derived from pESCs may enable in vitro modeling of early embryogenesis and improve livestock species' breeding practices.
Collapse
Affiliation(s)
- Jinzhu Xiang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Hanning Wang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Bingbo Shi
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Jiajun Li
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Dong Liu
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Kaipeng Wang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Zhuangfei Wang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Qiankun Min
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Chengchen Zhao
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
| | - Duanqing Pei
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Thiery JP, Sheng G, Shu X, Runyan R. How studies in developmental epithelial-mesenchymal transition and mesenchymal-epithelial transition inspired new research paradigms in biomedicine. Development 2024; 151:dev200128. [PMID: 38300897 DOI: 10.1242/dev.200128] [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] [Indexed: 02/03/2024]
Abstract
Epithelial-mesenchymal transition (EMT) and its reverse mechanism, mesenchymal-epithelial transition (MET), are evolutionarily conserved mechanisms initially identified in studies of early metazoan development. EMT may even have been established in choanoflagellates, the closest unicellular relative of Metazoa. These crucial morphological transitions operate during body plan formation and subsequently in organogenesis. These findings have prompted an increasing number of investigators in biomedicine to assess the importance of such mechanisms that drive epithelial cell plasticity in multiple diseases associated with congenital disabilities and fibrosis, and, most importantly, in the progression of carcinoma. EMT and MET also play crucial roles in regenerative medicine, notably by contributing epigenetic changes in somatic cells to initiate reprogramming into stem cells and their subsequent differentiation into distinct lineages.
Collapse
Affiliation(s)
| | - Guojun Sheng
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Xiaodong Shu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Raymond Runyan
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
4
|
Kajihara R, Ezaki R, Ichikawa K, Watanabe T, Terada T, Matsuzaki M, Horiuchi H. Wnt signaling blockade is essential for maintaining the pluripotency of chicken embryonic stem cells. Poult Sci 2024; 103:103361. [PMID: 38154448 PMCID: PMC10788285 DOI: 10.1016/j.psj.2023.103361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023] Open
Abstract
Activation of Wnt/β-catenin signaling supports the self-renewal of mouse embryonic stem cells. We aimed to understand the effects of Wnt signaling activation or inhibition on chicken embryonic stem cells (chESCs), as these effects are largely unknown. When the glycogen synthase kinase-3 β inhibitor CHIR99021-which activates Wnt signaling-was added to chESC cultures, the colony shape flattened, and the expression levels of pluripotency-related (NANOG, SOX2, SOX3, OCT4, LIN28A, DNMT3B, and PRDM14) and germ cell (CVH and DAZL) markers showed a decreasing trend, and the growth of chESCs was inhibited after approximately 7 d. By contrast, when the Wnt signaling inhibitor XAV939 was added to the culture, dense and compact multipotent colonies (morphologically similar to mouse embryonic stem cell colonies) showing stable expression of pluripotency-related and germline markers were formed. The addition of XAV939 stabilized the proliferation of chESCs in the early stages of culture and promoted their establishment. Furthermore, these chESCs formed chimeras. In conclusion, functional chESCs can be stably cultured using Wnt signaling inhibitors. These findings suggest the importance of Wnt/β-catenin signaling in avian stem cells, offering valuable insights for applied research using chESCs.
Collapse
Affiliation(s)
- Ryota Kajihara
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Ryo Ezaki
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Kennosuke Ichikawa
- The Roslin Institute, University of Edinburgh, Edinburgh EH25 9RG, United Kingdom
| | - Tenkai Watanabe
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Takumi Terada
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Mei Matsuzaki
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Hiroyuki Horiuchi
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima 739-8528, Japan; Genome Editing Innovation Center, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-0046, Japan.
| |
Collapse
|
5
|
Gabriel Francia M, Oses C, Lorena Roberti S, Reneé Garcia M, Helio Cozza L, Candelaria Diaz M, Levi V, Sonia Guberman A. SUMOylation and the oncogenic E17K mutation affect AKT1 subcellular distribution and impact on Nanog-binding dynamics to chromatin in embryonic stem cells. J Struct Biol 2023; 215:107961. [PMID: 37059313 DOI: 10.1016/j.jsb.2023.107961] [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: 12/26/2022] [Revised: 03/23/2023] [Accepted: 04/02/2023] [Indexed: 04/16/2023]
Abstract
AKT/PKB is a kinase involved in the regulation of a plethora of cell processes. Particularly, in embryonic stem cells (ESCs), AKT is crucial for the maintenance of pluripotency. Although the activation of this kinase relies on its recruitment to the cellular membrane and subsequent phosphorylation, multiple other post-translational modifications (PTMs), including SUMOylation, fine-tune its activity and target specificity. Since this PTM can also modify the localization and availability of different proteins, in this work we explored if SUMOylation impacts on the subcellular compartmentalization and distribution of AKT1 in ESCs. We found that this PTM does not affect AKT1 membrane recruitment, but it modifies the AKT1 nucleus/cytoplasm distribution, increasing its nuclear presence. Additionally, within this compartment, we found that AKT1 SUMOylation also impacts on the chromatin-binding dynamics of NANOG, a central pluripotency transcription factor. Remarkably, the oncogenic E17K AKT1 mutant produces major changes in all these parameters increasing the binding of NANOG to its targets, also in a SUMOylation dependent manner. These findings demonstrate that SUMOylation modulates AKT1 subcellular distribution, thus adding an extra layer of regulation of its function, possibly by affecting the specificity and interaction with its downstream targets.
Collapse
Affiliation(s)
- Marcos Gabriel Francia
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN, CONICET-UBA), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. Ciudad Autónoma de Buenos Aires, Argentina
| | - Camila Oses
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN, CONICET-UBA), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. Ciudad Autónoma de Buenos Aires, Argentina
| | - Sabrina Lorena Roberti
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN, CONICET-UBA), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. Ciudad Autónoma de Buenos Aires, Argentina
| | - Mora Reneé Garcia
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN, CONICET-UBA), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. Ciudad Autónoma de Buenos Aires, Argentina
| | - Lucas Helio Cozza
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN, CONICET-UBA), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. Ciudad Autónoma de Buenos Aires, Argentina
| | - Maria Candelaria Diaz
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN, CONICET-UBA), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. Ciudad Autónoma de Buenos Aires, Argentina
| | - Valeria Levi
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN, CONICET-UBA), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. Ciudad Autónoma de Buenos Aires, Argentina
| | - Alejandra Sonia Guberman
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN, CONICET-UBA), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. Ciudad Autónoma de Buenos Aires, Argentina; Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
6
|
Yoo DH, Im YS, Oh JY, Gil D, Kim YO. DUSP6 is a memory retention feedback regulator of ERK signaling for cellular resilience of human pluripotent stem cells in response to dissociation. Sci Rep 2023; 13:5683. [PMID: 37029196 PMCID: PMC10082014 DOI: 10.1038/s41598-023-32567-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 03/29/2023] [Indexed: 04/09/2023] Open
Abstract
Cultured human pluripotent stem cells (hPSCs) grow as colonies that require breakdown into small clumps for further propagation. Although cell death mechanism by single-cell dissociation of hPSCs has been well defined, how hPSCs respond to the deadly stimulus and recover the original status remains unclear. Here we show that dissociation of hPSCs immediately activates ERK, which subsequently activates RSK and induces DUSP6, an ERK-specific phosphatase. Although the activation is transient, DUSP6 expression persists days after passaging. DUSP6 depletion using the CRISPR/Cas9 system reveals that DUSP6 suppresses the ERK activity over the long term. Elevated ERK activity by DUSP6 depletion increases both viability of hPSCs after single-cell dissociation and differentiation propensity towards mesoderm and endoderm lineages. These findings provide new insights into how hPSCs respond to dissociation in order to maintain pluripotency.
Collapse
Affiliation(s)
- Dae Hoon Yoo
- Division of Intractable Disease Research, Korea National Institute of Health, Osong, Cheongju, 28160, Republic of Korea
| | - Young Sam Im
- Division of Intractable Disease Research, Korea National Institute of Health, Osong, Cheongju, 28160, Republic of Korea
| | - Ji Young Oh
- Division of Intractable Disease Research, Korea National Institute of Health, Osong, Cheongju, 28160, Republic of Korea
| | - Dayeon Gil
- Division of Intractable Disease Research, Korea National Institute of Health, Osong, Cheongju, 28160, Republic of Korea
| | - Yong-Ou Kim
- Division of Intractable Disease Research, Korea National Institute of Health, Osong, Cheongju, 28160, Republic of Korea.
- Center for National Stem Cell and Regenerative Medicine 202, Osongsaengmyung 2-Ro, Heundeok-Gu, Cheongju, Chungcheongbuk-Do, 28160, Republic of Korea.
| |
Collapse
|
7
|
Kalantary-Charvadeh A, Hosseini V, Mehdizadeh A, Nazari Soltan Ahmad S, Rahbarghazi R, Nozad Charoudeh H, Nouri M, Darabi M. The porcupine inhibitor WNT974 provokes ectodermal lineage differentiation of human embryonic stem cells. Cell Biochem Funct 2022; 40:359-368. [PMID: 35445405 DOI: 10.1002/cbf.3700] [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: 12/01/2021] [Revised: 03/04/2022] [Accepted: 03/10/2022] [Indexed: 11/08/2022]
Abstract
Porcupine (Porcn) enzyme plays an essential role in Wnt signaling activation. Stearoyl-CoA desaturase-1 (SCD1) is required to provide Porcn substrates. The aim of this study was to determine the effect of a novel Porcn inhibitor on the fate of human embryonic stem cells (hESCs) and the reliance of Porcn on SCD1 activity. hESCs were cultured on a feeder layer or Matrigel-coated plates. Small molecules WNT974 (LGK-974) and CAY10566 were used to inhibit Porcn and SCD1 activity, respectively. We assessed the effect of Porcn inhibition on viability, expression of Wnt signaling targets, pluripotency markers, proliferation, differentiation, and protein fatty acylation. hESCs' conditioned medium (CM) containing secreted Wnt proteins were applied in rescue experiments. To examine the catalytic dependency of Porcn on SCD1, the results of combined inhibitor treatment were compared with the SCD1 inhibitor alone. LGK-974 at the selected concentrations showed mild effects on hESCs viability, but significantly reduced messenger RNA and protein expression of Wnt signaling targets (Axin-2 and c-Myc) and pluripotency markers (OCT-4 and SOX-2) (p < .05). Adding 1 μM of Porcn inhibitor reduced proliferation (p = .03) and enhanced differentiation capacity into ectodermal progenitors (p = .02), which were reverted by CM. Click chemistry reaction did not show significant alteration in protein fatty acylation upon LGK-974 treatment. Moreover, combined inhibitor treatment caused no further substantial reduction in Wnt signaling targets, pluripotency markers, and protein fatty acylation relative to CAY10566-treated cultures. The substrate availability for Porcn activity is regulated by SCD1 and targeting Porcn by LGK-974 prompts the transition of hESCs from self-renewal state to ectodermal lineage.
Collapse
Affiliation(s)
- Ashkan Kalantary-Charvadeh
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Hosseini
- Molecular Medicine Research Center, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Nazari Soltan Ahmad
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hojjatollah Nozad Charoudeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Darabi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Molecular Cell Biology Laboratory, Internal Medicine IV, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
8
|
Pan T, Wang N, Zhang J, Yang F, Chen Y, Zhuang Y, Xu Y, Fang J, You K, Lin X, Li Y, Li S, Liang K, Li YX, Gao Y. Efficiently generate functional hepatic cells from human pluripotent stem cells by complete small-molecule strategy. Stem Cell Res Ther 2022; 13:159. [PMID: 35410439 PMCID: PMC8996222 DOI: 10.1186/s13287-022-02831-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Various methods have been developed to generate hepatic cells from human pluripotent stem cells (hPSCs) that rely on the combined use of multiple expensive growth factors, limiting industrial-scale production and widespread applications. Small molecules offer an attractive alternative to growth factors for producing hepatic cells since they are more economical and relatively stable. METHODS We dissect small-molecule combinations and identify the ideal cocktails to achieve an optimally efficient and cost-effective strategy for hepatic cells differentiation, expansion, and maturation. RESULTS We demonstrated that small-molecule cocktail CIP (including CHIR99021, IDE1, and PD0332991) efficiently induced definitive endoderm (DE) formation via increased endogenous TGF-β/Nodal signaling. Furthermore, we identified that combining Vitamin C, Dihexa, and Forskolin (VDF) could substitute growth factors to induce hepatic specification. The obtained hepatoblasts (HBs) could subsequently expand and mature into functional hepatocyte-like cells (HLCs) by the established chemical formulas. Thus, we established a stepwise strategy with complete small molecules for efficiently producing scalable HBs and functionally matured HLCs. The small-molecule-derived HLCs displayed typical functional characteristics as mature hepatocytes in vitro and repopulating injured liver in vivo. CONCLUSION Our current small-molecule-based hepatic generation protocol presents an efficient and cost-effective platform for the large-scale production of functional human hepatic cells for cell-based therapy and drug discovery using.
Collapse
Affiliation(s)
- Tingcai Pan
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou , Guangdong, China.,Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Ning Wang
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jiaye Zhang
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Fan Yang
- Guangdong Key Laboratory of Non-Human Primate Models, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong, China
| | - Yan Chen
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Yuanqi Zhuang
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Yingying Xu
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Ji Fang
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Kai You
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Xianhua Lin
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Yang Li
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou , Guangdong, China
| | - Shao Li
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou , Guangdong, China
| | - Kangyan Liang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou , Guangdong, China
| | - Yin-Xiong Li
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Yi Gao
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou , Guangdong, China. .,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Kannan P, Raghunathan M, Mohan T, Palanivelu S, Periandavan K. Gymnemic Acid Ameliorates Pancreatic β-Cell Dysfunction by Modulating Pdx1 Expression: A Possible Strategy for β-Cell Regeneration. Tissue Eng Regen Med 2022; 19:603-616. [PMID: 35212973 PMCID: PMC9130387 DOI: 10.1007/s13770-022-00435-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/08/2022] [Accepted: 01/19/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Endogenous pancreatic β-cell regeneration is a promising therapeutic approach for enhancing β-cell function and neogenesis in diabetes. Various findings have reported that regeneration might occur via stimulating β-cell proliferation, neogenesis, or conversion from other pancreatic cells to β-like cells. Although the current scenario illustrates numerous therapeutic strategies and approaches that concern endogenous β-cell regeneration, all of them have not been successful to a greater extent because of cost effectiveness, availability of suitable donors and rejection in case of transplantation, or lack of scientific evidence for many phytochemicals derived from plants that have been employed in traditional medicine. Therefore, the present study aims to investigate the effect of gymnemic acid (GA) on β-cell regeneration in streptozotocin-induced type 1 diabetic rats and high glucose exposed RIN5-F cells. METHODS The study involves histopathological and immunohistochemical analysis to examine the islet's architecture. Quantitative polymerase chain reaction (qPCR) and/or immunoblot were employed to quantify the β-cell regeneration markers and cell cycle proliferative markers. RESULTS The immunoexpression of E-cadherin, β-catenin, and phosphoinositide 3-kinases/protein kinase B were significantly increased in GA-treated diabetic rats. On the other hand, treatment with GA upregulated the pancreatic regenerative transcription factor viz. pancreatic duodenal homeobox 1, Neurogenin 3, MafA, NeuroD1, and β-cells proliferative markers such as CDK4, and Cyclin D1, with a simultaneous downregulation of the forkhead box O, glycogen synthase kinase-3, and p21cip1 in diabetic treated rats. Adding to this, we noticed increased nuclear localization of Pdx1 in GA treated high glucose exposed RIN5-F cells. CONCLUSION Our results suggested that GA acts as a potential therapeutic candidate for endogenous β-cell regeneration in treating type 1 diabetes.
Collapse
Affiliation(s)
- Pugazhendhi Kannan
- Department of Medical Biochemistry, Dr ALM PG IBMS, University of Madras, Taramani Campus, Taramani, Chennai, 600 113 India
| | - Malathi Raghunathan
- Department of Pathology, Dr ALM PG IBMS, University of Madras, Taramani Campus, Taramani, Chennai, India
| | - Thangarajeswari Mohan
- Department of Medical Biochemistry, Dr ALM PG IBMS, University of Madras, Taramani Campus, Taramani, Chennai, 600 113 India
| | - Shanthi Palanivelu
- Department of Pathology, Dr ALM PG IBMS, University of Madras, Taramani Campus, Taramani, Chennai, India
| | - Kalaiselvi Periandavan
- Department of Medical Biochemistry, Dr ALM PG IBMS, University of Madras, Taramani Campus, Taramani, Chennai, 600 113, India.
| |
Collapse
|
10
|
Yin Y, Shi L, Yang J, Wang H, Yang H, Wang Q. B7 family member H4 induces epithelial-mesenchymal transition and promotes the proliferation, migration and invasion of colorectal cancer cells. Bioengineered 2021; 13:107-118. [PMID: 34818980 PMCID: PMC8805878 DOI: 10.1080/21655979.2021.2009411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a common malignancy of the gastrointestinal tract, which has the second highest incidence among gastrointestinal tumors. At present, due to the limitations of current CRC treatment strategies, there is an urgent need for developing more effective therapies. B7 family member H4 (B7-H4) is associated with the progression of a wide spectrum of cancers, but its functional role in CRC is unknown. The purpose of this study is to clarify the role of B7-H4 in CRC and the underlying mechanisms in controlling the progression of CRC. Our data showed that B7-H4 expression in CRC tissues and cell lines was significantly upregulated as compared with normal tissues and normal cell lines. High B7-H4 expression was correlated with a poor prognosis of CRC patients. B7-H4 overexpression promoted the proliferation and invasion of CRC cells, which could be suppressed by Wnt signaling inhibitor. In a mouse xenograft model, silencing B7-H4 suppressed tumor growth and epithelial–mesenchymal transition (EMT) of CRC cells. Collectively, our study demonstrated the oncogenic roles of B7-H4 in regulating the proliferation, EMT as well as the migration of CRC cells through Wnt signaling pathway. The heightened expression of B7-H4 could serve as a prognostic marker for CRC patients.
Collapse
Affiliation(s)
- Yuzhen Yin
- Cancer Center, Jiangsu Shengze Hospital of Nanjing Medical University, Jiangsu, Suzhou, China
| | - Lili Shi
- Department of Ultrasound, Jiangsu Shengze Hospital of Nanjing Medical University, Jiangsu, Suzhou, China
| | - Jing Yang
- Cancer Center, Jiangsu Shengze Hospital of Nanjing Medical University, Jiangsu, Suzhou, China
| | - Hui Wang
- Department of Oncology, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Hang Yang
- Cancer Center, Jiangsu Shengze Hospital of Nanjing Medical University, Jiangsu, Suzhou, China
| | - Qiang Wang
- Department of General Surgery, Jiangsu Shengze Hospital of Nanjing Medical University, Jiangsu, Suzhou, China
| |
Collapse
|
11
|
Salerno AG, Wanschel ACBA, Dulce RA, Hatzistergos KE, Balkan W, Hare JM. S-nitrosoglutathione reductase (GSNOR) deficiency accelerates cardiomyocyte differentiation of induced pluripotent stem cells. THE JOURNAL OF CARDIOVASCULAR AGING 2021; 1. [PMID: 34790975 DOI: 10.20517/jca.2021.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Introduction Induced pluripotent stem cells (iPSCs) provide a model of cardiomyocyte (CM) maturation. Nitric oxide signaling promotes CM differentiation and maturation, although the mechanisms remain controversial. Aim The study tested the hypothesis that in the absence of S-nitrosoglutathione reductase (GSNOR), a denitrosylase regulating protein S-nitrosylation, the resultant increased S-nitrosylation accelerates the differentiation and maturation of iPSC-derived cardiomyocytes (CMs). Methods and Results iPSCs derived from mice lacking GSNOR (iPSCGSNOR-/-) matured faster than wildtype iPSCs (iPSCWT) and demonstrated transient increases in expression of murine Snail Family Transcriptional Repressor 1 gene (Snail), murine Snail Family Transcriptional Repressor 2 gene (Slug) and murine Twist Family BHLH Transcription Factor 1 gene (Twist), transcription factors that promote epithelial-to-mesenchymal transition (EMT) and that are regulated by Glycogen Synthase Kinase 3 Beta (GSK3β). Murine Glycogen Synthase Kinase 3 Beta (Gsk3β) gene exhibited much greater S-nitrosylation, but lower expression in iPSCGSNOR-/-. S-nitrosoglutathione (GSNO)-treated iPSCWT and human (h)iPSCs also demonstrated reduced expression of GSK3β. Nkx2.5 expression, a CM marker, was increased in iPSCGSNOR-/- upon directed differentiation toward CMs on Day 4, whereas murine Brachyury (t), Isl1, and GATA Binding Protein (Gata4) mRNA were decreased, compared to iPSCWT, suggesting that GSNOR deficiency promotes CM differentiation beginning immediately following cell adherence to the culture dish-transitioning from mesoderm to cardiac progenitor. Conclusion Together these findings suggest that increased S-nitrosylation of Gsk3β promotes CM differentiation and maturation from iPSCs. Manipulating the post-translational modification of GSK3β may provide an important translational target and offers new insight into understanding of CM differentiation from pluripotent stem cells. One sentence summary Deficiency of GSNOR or addition of GSNO accelerates early differentiation and maturation of iPSC-cardiomyocytes.
Collapse
Affiliation(s)
- Alessandro G Salerno
- Department of Medicine and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Amarylis C B A Wanschel
- Department of Medicine and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Raul A Dulce
- Department of Medicine and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Konstantinos E Hatzistergos
- Department of Medicine and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Wayne Balkan
- Department of Medicine and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua M Hare
- Department of Medicine and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
12
|
Ghuwalewala S, Ghatak D, Das S, Roy S, Das P, Butti R, Gorain M, Nath S, Kundu GC, Roychoudhury S. MiRNA-146a/AKT/β-Catenin Activation Regulates Cancer Stem Cell Phenotype in Oral Squamous Cell Carcinoma by Targeting CD24. Front Oncol 2021; 11:651692. [PMID: 34712602 PMCID: PMC8546321 DOI: 10.3389/fonc.2021.651692] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 09/16/2021] [Indexed: 01/06/2023] Open
Abstract
CD44highCD24low population has been previously reported as cancer stem cells (CSCs) in Oral Squamous Cell Carcinoma (OSCC). Increasing evidence suggests potential involvement of microRNA (miRNA) network in modulation of CSC properties. MiRNAs have thus emerged as crucial players in tumor development and maintenance. However, their role in maintenance of OSCC stem cells remains unclear. Here we report an elevated expression of miR-146a in the CD44highCD24low population within OSCC cells and primary HNSCC tumors. Moreover, over-expression of miR-146a results in enhanced stemness phenotype by augmenting the CD44highCD24low population. We demonstrate that miR-146a stabilizes β-catenin with concomitant loss of E-cadherin and CD24. Interestingly, CD24 is identified as a novel functional target of miR-146a and ectopic expression of CD24 abrogates miR-146a driven potential CSC phenotype. Mechanistic analysis reveals that higher CD24 levels inhibit AKT phosphorylation leading to β-catenin degradation. Using stably expressing miR-146a/CD24 OSCC cell lines, we also validate that the miR-146a/CD24/AKT loop significantly alters tumorigenic ability in vivo. Furthermore, we confirmed that β-catenin trans-activates miR-146a, thereby forming a positive feedback loop contributing to stem cell maintenance. Collectively, our study demonstrates that miR-146a regulates CSCs in OSCC through CD24-AKT-β-catenin axis.
Collapse
Affiliation(s)
- Sangeeta Ghuwalewala
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Dishari Ghatak
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Sumit Das
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune, India
| | - Stuti Roy
- Division of Research, Saroj Gupta Cancer Centre and Research Institute, Kolkata, India
| | - Pijush Das
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Ramesh Butti
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune, India
| | - Mahadeo Gorain
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune, India
| | - Somsubhra Nath
- Division of Research, Saroj Gupta Cancer Centre and Research Institute, Kolkata, India
| | - Gopal C Kundu
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune, India
| | - Susanta Roychoudhury
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Division of Research, Saroj Gupta Cancer Centre and Research Institute, Kolkata, India
| |
Collapse
|
13
|
Hosseini V, Kalantary-Charvadeh A, Hajikarami M, Fayyazpour P, Rahbarghazi R, Totonchi M, Darabi M. A small molecule modulating monounsaturated fatty acids and Wnt signaling confers maintenance to induced pluripotent stem cells against endodermal differentiation. Stem Cell Res Ther 2021; 12:550. [PMID: 34674740 PMCID: PMC8532309 DOI: 10.1186/s13287-021-02617-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/07/2021] [Indexed: 12/11/2022] Open
Abstract
Background Stearoyl-coenzyme A desaturase 1 (SCD1) is required for de novo synthesis of fatty acids. Through the fatty acid acylation process, this enzyme orchestrates post-translational modifications to proteins involved in cell development and differentiation. In this study, we used biochemical methods, immunostaining, and covalent labeling to evaluate whether a small molecule modulating unsaturated fatty acids can influence the early endodermal differentiation of human-induced pluripotent stem cells (iPSCs). Methods The hiPSCs were cultured in an endoderm-inducing medium containing activin A and defined fetal bovine serum in the presence of an SCD1 inhibitor at different time points. The cell cycles and the yields of the three germ layers (endoderm, mesoderm, and ectoderm) were assessed using flow cytometry. The expression of endoderm and pluripotency markers and the expressions of Wnt signaling pathway proteins were assessed using western blotting and RT-PCR. Total protein acylation was evaluated using a click chemistry reaction. Results When SCD1 was inhibited on the first day, the population of cells with endodermal features decreased at the end of differentiation. Moreover, early SCD1 inhibition preserved the properties of hiPSCs, preventing their shift toward mesodermal or ectodermal lineage. Also, first-day-only treatment of cells with the SCD1 inhibitor decreased β-catenin gene expression and the intensity of fluorescent emission in the click chemistry assay. The cells were effectively rescued from these effects by cotreatment with oleate. Late treatment with the inhibitor in the two subsequent days of endoderm induction did not have any significant effects on endoderm-specific markers or fluorescent intensity. Reproducible results were also obtained with human embryonic stem cells. Conclusion The small molecule SCD1 inhibitor attenuates the Wnt/β-catenin signaling pathway, conferring the maintenance of hiPSCs by opposing the initiation of endoderm differentiation. The immediate requirement for SCD1 activity in the endoderm commitment of pluripotent stem cells may be of importance in disorders of endoderm-derived organs and dysregulated metabolism. The schematic representation of the study design and main results. Activin A induces endoderm features through Smad2/3/4 and increases the expression of SCD1. SCD1 can produce MUFAs and subsequently modify the Wnt molecules. MUFA acylated/activated Wnts are secreted to interact with corresponding receptors on the target cells. β-catenin accumulates in the cytoplasm and is translocated into the nucleus after the interaction of Wnt with the receptor. Then, β-catenin increases the expression of the endoderm markers Sox17 and CXCR4.![]() Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02617-x.
Collapse
Affiliation(s)
- Vahid Hosseini
- Student Research Committee, Tabriz University of Medical Sciences, 5166615573, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, 516615731, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ashkan Kalantary-Charvadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Hajikarami
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Parisa Fayyazpour
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, 516615731, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Totonchi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Masoud Darabi
- Stem Cell Research Center, Tabriz University of Medical Sciences, 516615731, Tabriz, Iran. .,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Internal Medicine IV, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
14
|
Sun Y, Liu H, Sun XY, Xia W, Deng C. In vitro and in vivo study on the osseointegration of magnesium and strontium ion with two different proportions of mineralized collagen and its mechanism. J Biomater Appl 2021; 36:528-540. [PMID: 34000860 DOI: 10.1177/08853282211016934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
To explore the optimal combination of Mg2+, Sr2+ and mineralized collagen (nHAC) with two different proportions of hydroxyapatite (HA) and collagen (COL) on differentiation of MC3T3-E1 and the underlying mechanism, as well as achieve bone osseointegration. MC3T3-E1 cells were cultured in a complete medium with Mg2+ at the concentration of 0, 4, 8, 12, 16, 20 mmol/L, Sr2+ at the concentration of 0, 3, 6, 12 mmol/L, and the impregnation solution of 3:7 and 5:5nHAC. The differentiation of MC3T3-E1 was measured by expression of osteogenic genes and proteins including Runx-2, BMP-2 and OCN and determined the activation of PI3K/AKT/GSK3β/β-catenin signaling pathway in 12 mmol/LMg2++3 mmol/LSr2++3:7nHAC group. Osteoporosis was induced in 18 female rats by means of ovariectomy, the implants were immersed in 60 mmol/LMg2++15 mmol/LSr2++3:7nHAC impregnation solution and implanted into the mesial alveolar fossa for immediate implantation. The osseointegration of the implants was observed by Confocal laser scanning microscopy (CLSM) and histology at 4 and 8 weeks. The groups cultured with 12 mmol/LMg2+, 3 mmol/LSr2+ and 3:7nHAC impregnation solution showed the osteogenic genes and proteins were significantly higher respectively (P < 0.05), as well as p-Akt, p-GSK3β and β-catenin proteins (P < 0.05). CLSM and histology showed that the implant surface was surrounded by thick lamellar bone plate, and the trabecular bone were dense and continuous in the impregnation solution. These results found that magnesium and strontium ion-loaded mineralized collagen play an critical role in up-regulating the cells activity through PI3K/AKT/GSK3β/β-catenin signaling pathway and could be promote the formation of osseointegration.
Collapse
Affiliation(s)
- Yi Sun
- School of Stomatology, Wannan Medical College, WuHu, Anhui, PR China
| | - Hai Liu
- School of Stomatology, Wannan Medical College, WuHu, Anhui, PR China
| | - Xiao-Yu Sun
- School of Stomatology, Wannan Medical College, WuHu, Anhui, PR China
| | - Wen Xia
- School of Stomatology, Wannan Medical College, WuHu, Anhui, PR China
| | - Chao Deng
- School of Stomatology, Wannan Medical College, WuHu, Anhui, PR China
| |
Collapse
|
15
|
Zhu L, Hu S, Chen Q, Zhang H, Fu J, Zhou Y, Bai Y, Pan Y, Shao C. Macrophage contributes to radiation-induced anti-tumor abscopal effect on transplanted breast cancer by HMGB1/TNF-α signaling factors. Int J Biol Sci 2021; 17:926-941. [PMID: 33867819 PMCID: PMC8040298 DOI: 10.7150/ijbs.57445] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Objectives: The roles of innate immunity including macrophages in radiation-induced abscopal effect (RIAE) are ambiguous. In this study, we evaluated the role of macrophage in RIAE and the interaction of cytokines in tumor microenvironment after irradiation. Materials and Methods: Transplanted tumor of breast cancer cells in BalB/C mice, severe combined immunodeficiency (SCID) mice and non-obese diabetic (NOD)-SCID mice were irradiated with fractionation doses to observe anti-tumor abscopal effect. The underlying mechanism of RIAE was investigated by treating the mice with TNF-α inhibitor or macrophage depletion drug and analyzing the alteration of macrophage distribution in tumors. A co-culture system of breast cancer cells and macrophages was applied to disclose the signaling factors and related pathways involved in the RIAE. Results: The growth of nonirradiated tumor was effectively suppressed in mice with normal or infused macrophages but not in mice with insufficiency/depletion of macrophage or TNF-α inhibition, where M1-macrophage was mainly involved. Investigation of the bystander signaling factors in vitro demonstrated that HMGB1 released from irradiated breast cancer cells promoted bystander macrophages to secret TNF-α through TLR-4 pathway and further inhibited the proliferation and migration of non-irradiated cancer cells by PI3K-p110γ suppression. Conclusions: HMGB1 and TNF-α contributes to M1-macrophages facilitated systemic anti-tumor abscopal response triggered by radiotherapy in breast cancer, indicating that the combination of immunotherapy and radiotherapy may has important implication in enhancing the efficiency of tumor treatment.
Collapse
Affiliation(s)
- Lin Zhu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Songling Hu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Qianping Chen
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Haowen Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China.,State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China
| | - Jiamei Fu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Yuchuan Zhou
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yang Bai
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yan Pan
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
16
|
McKee C, Brown C, Bakshi S, Walker K, Govind CK, Chaudhry GR. Transcriptomic Analysis of Naïve Human Embryonic Stem Cells Cultured in Three-Dimensional PEG Scaffolds. Biomolecules 2020; 11:E21. [PMID: 33379237 PMCID: PMC7824559 DOI: 10.3390/biom11010021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/09/2020] [Accepted: 12/24/2020] [Indexed: 12/21/2022] Open
Abstract
Naïve human embryonic stem cells (ESCs) are characterized by improved viability, proliferation, and differentiation capacity in comparison to traditionally derived primed human ESCs. However, currently used two-dimensional (2-D) cell culture techniques fail to mimic the three-dimensional (3-D) in vivo microenvironment, altering morphological and molecular characteristics of ESCs. Here, we describe the use of 3-D self-assembling scaffolds that support growth and maintenance of the naïve state characteristics of ESC line, Elf1. Scaffolds were formed via a Michael addition reaction upon the combination of two 8-arm polyethylene glycol (PEG) polymers functionalized with thiol (PEG-8-SH) and acrylate (PEG-8-Acr) end groups. 3-D scaffold environment maintained the naïve state and supported the long-term growth of ESCs. RNA-sequencing demonstrated significant changes in gene expression profiles between 2-D and 3-D grown cells. Gene ontology analysis revealed upregulation of biological processes involved in the regulation of transcription and translation, extracellular matrix organization, and chromatin remodeling in 3-D grown cells. 3-D culture conditions also induced upregulation of genes associated with Wnt and focal adhesion signaling, while p53 signaling pathway associated genes were downregulated. Our findings, for the first time, provide insight into the possible mechanisms of self-renewal of naïve ESCs stimulated by the transduction of mechanical signals from the 3-D microenvironment.
Collapse
Affiliation(s)
- Christina McKee
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - Christina Brown
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - Shreeya Bakshi
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - Keegan Walker
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - Chhabi K. Govind
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - G. Rasul Chaudhry
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| |
Collapse
|
17
|
Kalantary-Charvadeh A, Hosseini V, Mehdizadeh A, Darabi M. Application of porcupine inhibitors in stem cell fate determination. Chem Biol Drug Des 2020; 96:1052-1068. [PMID: 32419352 DOI: 10.1111/cbdd.13704] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 04/27/2020] [Accepted: 05/03/2020] [Indexed: 02/06/2023]
Abstract
Porcupine (Porcn), a membrane-bound O-acyltransferase, is an endoplasmic reticulum-located protein that has catalytic activity. Porcn is involved in post-translational lipid modification of wingless-Int (Wnt) proteins and serves as an indispensable step in the Wnt proper secretion and signaling. Small-molecule inhibitors targeting Porcn catalytic function in vitro and in vivo are of great interest not only for treating cancer and fibrotic disorders but also in the field of regenerative medicine. Although a number of studies have been conducted, the exact role of Porcn in stem cell fate is not entirely clear. In some cases, Porcn inhibition declined differentiation rate, and in others, it induced stem cell differentiation toward specific lineages. In this review, we first elaborated the Porcn catalytic activity and its inhibitors. Then, we discussed about the recently reported results of Porcn inhibitors in stem cells self-renewal and differentiation.
Collapse
Affiliation(s)
- Ashkan Kalantary-Charvadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Hosseini
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Darabi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
18
|
Menezes SV, Fouani L, Huang MLH, Geleta B, Maleki S, Richardson A, Richardson DR, Kovacevic Z. The metastasis suppressor, NDRG1, attenuates oncogenic TGF-β and NF-κB signaling to enhance membrane E-cadherin expression in pancreatic cancer cells. Carcinogenesis 2020; 40:805-818. [PMID: 30561520 DOI: 10.1093/carcin/bgy178] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/18/2018] [Accepted: 12/13/2018] [Indexed: 01/04/2023] Open
Abstract
The metastasis suppressor, N-myc downstream-regulated gene-1 (NDRG1), plays multifaceted roles in inhibiting oncogenic signaling and can suppress the epithelial mesenchymal transition (EMT), a key step in metastasis. In this investigation, NDRG1 inhibited the oncogenic effects of transforming growth factor-β (TGF-β) in PANC-1 pancreatic cancer cells, promoting expression and co-localization of E-cadherin and β-catenin at the cell membrane. A similar effect of NDRG1 at supporting E-cadherin and β-catenin co-localization at the cell membrane was also demonstrated for HT-29 colon and CFPAC-1 pancreatic cancer cells. The increase in E-cadherin in PANC-1 cells in response to NDRG1 was mediated by the reduction of three transcriptional repressors of E-cadherin, namely SNAIL, SLUG and ZEB1. To dissect the mechanisms how NDRG1 inhibits nuclear SNAIL, SLUG and ZEB1, we assessed involvement of the nuclear factor-κB (NF-κB) pathway, as its aberrant activation contributes to the EMT. Interestingly, NDRG1 comprehensively inhibited oncogenic NF-κB signaling at multiple sites in this pathway, suppressing NEMO, Iĸĸα and IĸBα expression, as well as reducing the activating phosphorylation of Iĸĸα/β and IĸBα. NDRG1 also reduced the levels, nuclear co-localization and DNA-binding activity of NF-κB p65. Further, Iĸĸα, which integrates NF-κB and TGF-β signaling to upregulate ZEB1, SNAIL and SLUG, was identified as an NDRG1 target. Considering this, therapies targeting NDRG1 could be a new strategy to inhibit metastasis, and as such, we examined novel anticancer agents, namely di-2-pyridylketone thiosemicarbazones, which upregulate NDRG1. These agents downregulated SNAIL, SLUG and ZEB1 in vitro and in vivo using a PANC-1 tumor xenograft model, demonstrating their marked potential.
Collapse
Affiliation(s)
- Sharleen V Menezes
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Leyla Fouani
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Michael L H Huang
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Bekesho Geleta
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Sanaz Maleki
- Histopathology Laboratory, School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Alexander Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia.,Department of Pathology and Biological Responses, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Zaklina Kovacevic
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
19
|
Shen H, Ding C, Yuan S, Pan T, Li D, Li H, Huang B, Liu Q. Vitamin C- and Valproic Acid-Induced Fetal RPE Stem-like Cells Recover Retinal Degeneration via Regulating SOX2. Mol Ther 2020; 28:1645-1657. [PMID: 32353323 DOI: 10.1016/j.ymthe.2020.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 02/19/2020] [Accepted: 04/09/2020] [Indexed: 02/06/2023] Open
Abstract
Retinal pigment epithelial (RPE) cell replacement therapy has provided promising outcomes in the treatment of retinal degenerative diseases (RDDs), but the resulting limited visual improvement has raised questions about graft survival and differentiation. Through combined treatment with vitamin C and valproic acid (together, VV), we activated human fetal RPE (fRPE) cells to become highly proliferative fetal RPE stem-like cells (fRPESCs). In this study, we report that SOX2 (SRY-box 2) activation contributed to mesenchymal-epithelial transition and elevated the retinal progenitor and mesenchymal stromal markers expressions of fRPESCs. These fRPESCs could differentiate into RPE cells, rod photoreceptors, and mesenchymal lineage progenies under defined conditions. Finally, fRPESCs were transplanted into the subretinal space of an RDD mouse model, and a photoreceptor rescue benefit was demonstrated. The RPE and rod photoreceptor differentiation of transplanted fRPESCs may account for the neural retinal recovery. This study establishes fRPESCs as a highly proliferative, multi-lineage differentiation potential (including RPE, rod photoreceptor, and mesenchymal lineage differentiation), mesenchymal-to-epithelial-transitioned retinal stem-like cell source for cell-based therapy of RDDs.
Collapse
Affiliation(s)
- Han Shen
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chenyue Ding
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215002, China
| | - Songtao Yuan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ting Pan
- The Affiliated Changzhou No. 2 People's Hospital with Nanjing Medical University, Changzhou 213000, China
| | - Duo Li
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hong Li
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215002, China
| | - Boxian Huang
- Center of Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215002, China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, China.
| | - Qinghuai Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
20
|
Elucidating the mechanism of action of alpha-1-antitrypsin using retinal pigment epithelium cells exposed to high glucose. Potential use in diabetic retinopathy. PLoS One 2020; 15:e0228895. [PMID: 32032388 PMCID: PMC7006930 DOI: 10.1371/journal.pone.0228895] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/24/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alpha-1-antitrypsin is a protein involved in avoidance of different processes that are seen in diabetic retinopathy pathogenesis. These processes include apoptosis, extracellular matrix remodeling and damage of vessel walls and capillaries. Furthermore, because of its anti-inflammatory effects, alpha-1-antitrypsin has been proposed as a possible therapeutic approach for diabetic retinopathy. Our group tested alpha-1-antitrypsin in a type 1 diabetes mouse model and observed a reduction of inflammation and retinal neurodegeneration. Thus, shedding light on the mechanism of action of alpha-1-antitrypsin at molecular level may explain how it works in the diabetic retinopathy context and show its potential for use in other retinal diseases. METHODS In this work, we evaluated alpha-1-antitrypsin in an ARPE-19 human cell line exposed to high glucose. We explored the expression of different mediators on signaling pathways related to pro-inflammatory cytokines production, glucose metabolism, epithelial-mesenchymal transition and other proteins involved in the normal function of retinal pigment epithelium by RT-qPCR and Western Blot. RESULTS We obtained different expression patterns for evaluated mediators altered with high glucose exposure and corrected with the use of alpha-1-antitrypsin. CONCLUSIONS The expression profile obtained in vitro for the evaluated proteins and mRNA allowed us to explain our previous results obtained on mouse models and to hypothesize how alpha-1-antitrypsin hinder diabetic retinopathy progression on a complex network between different signaling pathways. GENERAL SIGNIFICANCE This network helps to understand the way alpha-1-antitrypsin works in diabetic retinopathy and its scope of action.
Collapse
|
21
|
Li Y, Wu S, Li X, Guo S, Cai Z, Yin Z, Zhang Y, Liu Z. Wnt signaling associated small molecules improve the viability of pPSCs in a PI3K/Akt pathway dependent way. J Cell Physiol 2020; 235:5811-5822. [DOI: 10.1002/jcp.29514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 01/08/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Yan Li
- Laboratory of Embryo BiotechnologyCollege of Life Science, Northeast Agricultural UniversityHarbin China
| | - Shuang Wu
- Laboratory of Embryo BiotechnologyCollege of Life Science, Northeast Agricultural UniversityHarbin China
| | - Xuechun Li
- Laboratory of Embryo BiotechnologyCollege of Life Science, Northeast Agricultural UniversityHarbin China
| | - Shimeng Guo
- Laboratory of Embryo BiotechnologyCollege of Life Science, Northeast Agricultural UniversityHarbin China
| | - Zhuang Cai
- Laboratory of Embryo BiotechnologyCollege of Life Science, Northeast Agricultural UniversityHarbin China
| | - Zhi Yin
- Laboratory of Embryo BiotechnologyCollege of Life Science, Northeast Agricultural UniversityHarbin China
| | - Yu Zhang
- Laboratory of Embryo BiotechnologyCollege of Life Science, Northeast Agricultural UniversityHarbin China
| | - Zhonghua Liu
- Laboratory of Embryo BiotechnologyCollege of Life Science, Northeast Agricultural UniversityHarbin China
| |
Collapse
|
22
|
Ye T, Li J, Sun Z, Liu D, Zeng B, Zhao Q, Wang J, Xing HR. Cdh1 functions as an oncogene by inducing self-renewal of lung cancer stem-like cells via oncogenic pathways. Int J Biol Sci 2020; 16:447-459. [PMID: 32015681 PMCID: PMC6990901 DOI: 10.7150/ijbs.38672] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/04/2019] [Indexed: 01/01/2023] Open
Abstract
The mortality rate of lung cancer remains the highest amongst all cancers despite of new therapeutic developments. While cancer stem cells (CSCs) may play a pivotal role in cancer, mechanisms underlying CSCs self-renewal and their relevance to cancer progression have not been clearly elucidated due to the lack of reliable and stable CSC cellular models. In the present study, we unveiled the novel oncogene function of cadherin 1 (Cdh1) via bioinformatic analysis in a broad spectrum of human cancers including lung adenocarcinoma (LUAD), adding a new dimension to the widely reported tumor suppressor function of Cdh1. Experimentally, we show for the first time that Cdh1 promotes the self-renewal of lung CSCs, consistent with its function in embryonic and normal stem cells. Using the LLC-Symmetric Division (LLC-SD) model, we have revealed an intricate cross-talk between the oncogenic pathway and stem cell pathway in which Cdh1 functions as an oncogene by promoting lung CSC renewal via the activation of the Phosphoinositide 3-kinase (PI3K) and inhibition of Mitogen-activated protein kinase (MAPK) pathways, respectively. In summary, this study has provided evidence demonstrating effective utilization of the normal stem cell renewal mechanisms by CSCs to promote oncogenesis and progression.
Collapse
Affiliation(s)
- Ting Ye
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China.,Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jingyuan Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China
| | - Zhiwei Sun
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China
| | - Doudou Liu
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China
| | - Bin Zeng
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China
| | - Qiting Zhao
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China
| | - Jianyu Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.,Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China
| | - H Rosie Xing
- Laboratory of Translational Cancer Stem Cell Research, Chongqing Medical University, Chongqing, China.,College of Biomedical Engineering, State Key Laboratory of Ultrasound Engineering in Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
23
|
朱 云, 李 成, 林 鑫, 孙 晶, 程 旸. [Effect of Atractylodes macrocephala polysaccharide on proliferation and invasion of hepatocellular carcinoma cells in vitro]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:1180-1185. [PMID: 31801717 PMCID: PMC6867946 DOI: 10.12122/j.issn.1673-4254.2019.10.08] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To investigate the inhibitory effect of polysaccharide of Atractylodes macrocephala (PAM) on the proliferation and invasion of hepatocellular carcinoma cells and the underlying mechanism. METHODS Hepatocellular carcinoma HepG2 cells were treated with different concentrations of PAM, and their proliferation and invasive ability were examined using CCK-8 assay and Transwell assay. Immunofluorescence assay was performed to detect the expression level of β-catenin, and real-time PCR and Western blotting were used to detect the mRNA and protein expressions of AKT, GSK-3β and MMP-2 in the cells. The changes in the proliferation, invasiveness and the expressions of pGSK-3β and MMP2 were examined in the cells following treatment with LiCl/PAM/LiCl plus PAM. RESULTS PAM treatment significantly reduced the cell viability, the number of migration cells, and the expression levels of β-catenin and MMP-2 (P < 0.05), and obviously inhibited the phosphorylation of AKT and GSK-3β in the cells (P < 0.05) in a dose-dependent manner. The rescue experiment showed that LiCl reversed the inhibition of cell proliferation, invasiveness, and the Wnt/β-catenin pathway induced by PAM. CONCLUSIONS PAM can inhibit the proliferation and invasion of hepatocellular carcinoma cells in vitro possibly by inhibiting the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- 云 朱
- 南方医科大学南方医院感染内科肝脏肿瘤中心,广东 广州 510515Liver Tumor Center, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, Guangzgou 510515, China
| | - 成 李
- 南方医科大学第二临床医学院,广东 广州 510280The Second Clinical Medical College, Southern Medical University, Guangzhou, 510280, China
| | - 鑫盛 林
- 南方医科大学第二临床医学院,广东 广州 510280The Second Clinical Medical College, Southern Medical University, Guangzhou, 510280, China
| | - 晶晶 孙
- 广州医科大学附属广州市妇女儿童医疗中心消化科,广东 广州 510623Digestive Department, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - 旸 程
- 广州医科大学附属广州市妇女儿童医疗中心消化科,广东 广州 510623Digestive Department, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| |
Collapse
|
24
|
Han U, Kim YJ, Kim W, Park JH, Hong J. Construction of nano-scale cellular environments by coating a multilayer nanofilm on the surface of human induced pluripotent stem cells. NANOSCALE 2019; 11:13541-13551. [PMID: 31290516 DOI: 10.1039/c9nr02375e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Interactions with peripheral environments, such as extracellular matrix (ECM) and other cells, and their balance play a crucial role in the maintenance of pluripotency and self-renewal of human pluripotent stem cells. In this study, we focused on a nano-sized artificial cellular environment that is directly attached to the cytoplasmic membrane as a facile method that can effect intercellular interactions at the single-cell level. We designed multilayered nanofilms that are self-assembled on the surface of human induced pluripotent stem cells (iPSCs), by repetitive adsorption of fibronectin and heparin or chondroitin sulfate. However, the surface modification process could also lead to the loss of cell-cell adhesion, which may result in apoptotic cell death. We investigated the proliferation and pluripotency of the iPSCs coated with the nanofilm in order to establish the suitable nanofilm structure and coating conditions. As a result, the cell viability reduced with the increase in the duration of the coating process, but the undifferentiated state and proliferation of the cells were maintained until 2 bilayers were coated. To suppress the dissociation-induced apoptosis, Y-27632, the Rho-associated kinase inhibitor (ROCKi), was added to the coating solution; this allowed the coating of up to 4 bilayers of the nanofilm onto the iPSCs. These results are expected to accelerate the pace of iPSC studies on 3-dimensional cultures and naïve pluripotency, in which the regulation of cellular interactions plays a critical role.
Collapse
Affiliation(s)
- Uiyoung Han
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| | - Yu Jin Kim
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon, Gangwon-do 24341, Republic of Korea.
| | - Wijin Kim
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon, Gangwon-do 24341, Republic of Korea.
| | - Ju Hyun Park
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon, Gangwon-do 24341, Republic of Korea.
| | - Jinkee Hong
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| |
Collapse
|
25
|
Three-dimensional cultured liver-on-a-Chip with mature hepatocyte-like cells derived from human pluripotent stem cells. Biomed Microdevices 2019; 21:73. [DOI: 10.1007/s10544-019-0423-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
26
|
Mitochondrial Akt Signaling Modulated Reprogramming of Somatic Cells. Sci Rep 2019; 9:9919. [PMID: 31289326 PMCID: PMC6616364 DOI: 10.1038/s41598-019-46359-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/27/2019] [Indexed: 12/13/2022] Open
Abstract
The signaling mechanisms controlling somatic cell reprogramming are not fully understood. In this study, we report a novel role for mitochondrial Akt1 signaling that enhanced somatic cell reprogramming efficiency. The role of mitochondrial Akt1 in somatic cell reprogramming was investigated by transducing fibroblasts with the four reprogramming factors (Oct4, Sox2, Klf4, c-Myc) in conjunction with Mito-Akt1, Mito-dnAkt1, or control virus. Mito-Akt1 enhanced reprogramming efficiency whereas Mito-dnAkt1 inhibited reprogramming. The resulting iPSCs formed embryoid bodies in vitro and teratomas in vivo. Moreover, Oct4 and Nanog promoter methylation was reduced in the iPSCs generated in the presence of Mito-Akt1. Akt1 was activated and translocated into mitochondria after growth factor stimulation in embryonic stem cells (ESCs). To study the effect of mitochondrial Akt in ESCs, a mitochondria-targeting constitutively active Akt1 (Mito-Akt1) was expressed in ESCs. Gene expression profiling showed upregulation of genes that promote stem cell proliferation and survival and down-regulation of genes that promote differentiation. Analysis of cellular respiration indicated similar metabolic profile in the resulting iPSCs and ESCs, suggesting comparable bioenergetics. These findings showed that activation of mitochondrial Akt1 signaling was required during somatic cell reprogramming.
Collapse
|
27
|
Zhang C, Zhang M, Ge S, Huang W, Lin X, Gao J, Gong J, Shen L. Reduced m6A modification predicts malignant phenotypes and augmented Wnt/PI3K-Akt signaling in gastric cancer. Cancer Med 2019; 8:4766-4781. [PMID: 31243897 PMCID: PMC6712480 DOI: 10.1002/cam4.2360] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/14/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
Background As the most abundant epigenetic modification on mRNAs and long non‐coding RNAs, N6‐methyladenosine (m6A) modification extensively exists in mammalian cells. Controlled by writers (methyltransferases), readers (signal transducers), and erasers (demethylases), m6A influences mRNA structure, maturation, and stability, thus negatively regulating protein expression in a post‐translational manner. Nevertheless, current understanding of m6A's roles in tumorigenesis, especially in gastric cancer (GC) remains to be unveiled. In this study, we assessed m6A's clinicopathological relevance to GC and explored the underlying mechanisms. Methods By referring to a proteomics‐based GC cohort we previously generated and the TCGA‐GC cohort, we merged expressions of canonical m6A writers (METTL3/METTL14), readers (YTHDF1/YTHDF2/YTHDF3), and erasers (ALKBH5/FTO), respectively, as W, R, and E signatures to represent m6A modification. We stratified patients according to these signatures to decipher m6A's associations with crucial mutations, prognosis, and clinical indexes. m6A's biological functions in GC were predicted by gene set enrichment analysis (GSEA) and validated by in vitro experiments. Results We discovered that W and R were potential tumor suppressive signatures, while E was a potential oncogenic signature in GC. According to W/R/E stratifications, patients with low m6A‐indications were accompanied with higher mutations of specific genes (CDH1, AR, GLI3, SETBP1, RHOA, MUC6, and TP53) and also demonstrated adverse clinical outcomes. GSEA suggested that reduced m6A was correlated with oncogenic signaling and phenotypes. Through in vitro experiments, we proved that m6A suppression (represented by METTL14 knockdown) promoted GC cell proliferation and invasiveness through activating Wnt and PI3K‐Akt signaling, while m6A elevation (represented by FTO knockdown) reversed these phenotypical and molecular changes. m6A may also be involved in interferon signaling and immune responses of GC. Conclusions Our work demonstrated that low‐m6A signatures predicted adverse clinicopathological features of GC, while the reduction of RNA m6A methylation activated oncogenic Wnt/PI3K‐Akt signaling and promoted malignant phenotypes of GC cells.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Mengqi Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Sai Ge
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Wenwen Huang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaoting Lin
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jing Gao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
28
|
Roy A, Murphy RM, Deng M, MacDonald JW, Bammler TK, Aldinger KA, Glass IA, Millen KJ. PI3K-Yap activity drives cortical gyrification and hydrocephalus in mice. eLife 2019; 8:45961. [PMID: 31094678 PMCID: PMC6544437 DOI: 10.7554/elife.45961] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/15/2019] [Indexed: 01/07/2023] Open
Abstract
Mechanisms driving the initiation of brain folding are incompletely understood. We have previously characterized mouse models recapitulating human PIK3CA-related brain overgrowth, epilepsy, dysplastic gyrification and hydrocephalus (Roy et al., 2015). Using the same, highly regulatable brain-specific model, here we report PI3K-dependent mechanisms underlying gyrification of the normally smooth mouse cortex, and hydrocephalus. We demonstrate that a brief embryonic Pik3ca activation was sufficient to drive subtle changes in apical cell adhesion and subcellular Yap translocation, causing focal proliferation and subsequent initiation of the stereotypic ‘gyrification sequence’, seen in naturally gyrencephalic mammals. Treatment with verteporfin, a nuclear Yap inhibitor, restored apical surface integrity, normalized proliferation, attenuated gyrification and rescued the associated hydrocephalus, highlighting the interrelated role of regulated PI3K-Yap signaling in normal neural-ependymal development. Our data defines apical cell-adhesion as the earliest known substrate for cortical gyrification. In addition, our preclinical results support the testing of Yap-related small-molecule therapeutics for developmental hydrocephalus.
Collapse
Affiliation(s)
- Achira Roy
- Center for Integrative Brain Research, Seattle Children's Research Institute, Washington, United States
| | - Rory M Murphy
- Center for Integrative Brain Research, Seattle Children's Research Institute, Washington, United States
| | - Mei Deng
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Washington, United States
| | - James W MacDonald
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Washington, United States
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Washington, United States
| | - Kimberly A Aldinger
- Center for Integrative Brain Research, Seattle Children's Research Institute, Washington, United States.,Division of Genetic Medicine, Department of Pediatrics, University of Washington, Washington, United States
| | - Ian A Glass
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Washington, United States
| | - Kathleen J Millen
- Center for Integrative Brain Research, Seattle Children's Research Institute, Washington, United States.,Division of Genetic Medicine, Department of Pediatrics, University of Washington, Washington, United States
| |
Collapse
|
29
|
Timmermans-Sprang E, Collin R, Henkes A, Philipsen M, Mol JA. P-cadherin mutations are associated with high basal Wnt activity and stemness in canine mammary tumor cell lines. Oncotarget 2019; 10:2930-2946. [PMID: 31105876 PMCID: PMC6508207 DOI: 10.18632/oncotarget.26873] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 04/04/2019] [Indexed: 01/16/2023] Open
Abstract
Purpose: To find underlying mutations causing highly-activated Wnt activity in mammary tumor cell lines associated with rounded morphology indicative of stemness/EMT. Methods: Stemness of high Wnt cell lines was confirmed using qPCR on selected genes and microRNA profiling, followed by whole-exome sequencing of 3 high Wnt canine mammary tumor cell lines and 5 low/absent Wnt cell lines. Candidate genes were identified and their involvement in Wnt activity investigated using siRNA silencing. Results: The high Wnt cell lines had morphological and gene expression characteristics reminiscent of stemness. All individual cell lines had about 4000 mutations in the exome in comparison to the reference canine genome. The three high basal Wnt cell lines had 167 unique exome mutations. Seven of these mutations resulted in a SIFT score <0.2 of proteins related to Wnt signaling. However, gene silencing did not change the Wnt pathway activation. Renewed analysis with respect to putative relations to Wnt signaling revealed that P-cadherin (CDH3) had three mutations in the coding region of the extracellular domain and was associated with high Wnt signaling. Silencing by siRNA not only in lowered Wnt activity, but also decreased levels of phosphorylated cSRC and sP-cad, and changed cell morphology towards spindle cell appearance. Conclusion: It is concluded that expression of mutated CDH3 is associated with activation of cSRC, stabilization of ß-catenin and a rounded morphology related to a stemness/EMT phenotype. A decreased Wnt activity can be found also by cSRC inhibition, but CDH3 silencing has an additional effect on morphology indicating reversal of EMT.
Collapse
Affiliation(s)
- Elpetra Timmermans-Sprang
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Rob Collin
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arjen Henkes
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Meike Philipsen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jan A. Mol
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
30
|
Zhang T, Chen H, Zhou Y, Dong W, Cai H, Tan WS. Cooperation of FGF/MEK/ERK and Wnt/β-catenin pathway regulators to promote the proliferation and pluripotency of mouse embryonic stem cells in serum- and feeder-free conditions. BIORESOUR BIOPROCESS 2019. [DOI: 10.1186/s40643-019-0249-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
31
|
Yu S, Zhang Y, Wu Y, Yang H, Chen Y, Yang Y, Zhang Z. Subcellular localization of mutated β-catenins with different incidences of cis-peptide bonds at the Xaa246-P247 site in HepG2 cells. FASEB J 2019; 33:6574-6583. [PMID: 30807699 PMCID: PMC6497428 DOI: 10.1096/fj.201801937rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Mutations may ultimately change the local conformation of proteins; however, little attention has been paid to alterations in protein function caused by the incidence of cis-peptide bonds (ICPB) in mammalian cells. In this study, a statistical approach, coimmunoprecipitation, and immunofluorescence staining have been used to confirm that S246→Y and S246→W missense mutations, which help increase the ICPB in Xaa246-P247 (Xaa is any amino acid) in human β-catenin, can reduce the interactions between β-catenin and adenomatous polyposis coli (APC) and between β-catenin and Ca2+-dependent cell adhesion molecule family in epithelial tissue (E-cadherin), eventually leading to increased nuclear migration of β-catenin in the HepG2 cell line (an immortalized cell line consisting of human liver carcinoma cells). Conversely, S246→L and S246→M missense mutations, which reduce the ICPB in Xaa246-P247 in human β-catenin, can enhance interactions between β-catenin and APC and between β-catenin and E-cadherin, leading to decreased nuclear migration of β-catenin. These results not only indicate that a change in the ICPB may be an important cause of functional protein changes but also provide a new basis for the study of genetic disease prediction, gene diagnosis, individualized treatment, and protein modification at the gene level for clinicians and other professionals.—Yu, S., Zhang, Y., Wu, Y., Yang, H., Chen, Y., Yang, Y., Zhang, Z. Subcellular localization of mutated β-catenins with different incidences of cis-peptide bonds at the Xaa246-P247 site in HepG2 cells.
Collapse
Affiliation(s)
- Shuhui Yu
- School of Life Sciences, Chongqing University, Chongqing, China.,Information Institute of Southwest University, Southwest University, Chongqing, China
| | - Yali Zhang
- School of Life Science, Southwest University, Chongqing, China.,Department of Hematology, First Affiliated Hospital of Army Military Medical University, Chongqing, China
| | - Yuyun Wu
- School of Life Science, Southwest University, Chongqing, China
| | - Hongying Yang
- School of Life Science, Southwest University, Chongqing, China
| | - Yang Chen
- School of Life Science, Southwest University, Chongqing, China
| | - Yingbin Yang
- School of Life Science, Southwest University, Chongqing, China
| | - Ze Zhang
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
32
|
Abstract
The organs require oxygen and other types of nutrients (amino acids, sugars, and lipids) to function, the heart consuming large amounts of fatty acids for oxidation and adenosine triphosphate (ATP) generation.
Collapse
|
33
|
Kovacic B, Rosner M, Schlangen K, Kramer N, Hengstschläger M. DRUGPATH - a novel bioinformatic approach identifies DNA-damage pathway as a regulator of size maintenance in human ESCs and iPSCs. Sci Rep 2019; 9:1897. [PMID: 30760778 PMCID: PMC6374489 DOI: 10.1038/s41598-018-37491-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 11/29/2018] [Indexed: 02/05/2023] Open
Abstract
Genetic and biochemical screening approaches often fail to identify functionally relevant pathway networks because many signaling proteins contribute to multiple gene ontology pathways. We developed a DRUGPATH-approach to predict pathway-interactomes from high-content drug screen data. DRUGPATH is based upon combining z-scores of effective inhibitors with their corresponding and validated targets. We test DRUGPATH by comparing homeostatic pathways in human embryonic stem cells (hESCs), human induced pluripotent stem cells (hiPSCs) and human amniotic fluid stem cells (hAFSCs). We show that hAFSCs utilize distinct interactomes compared to hESCs/hiPSCs and that pathways orchestrating cell cycle and apoptosis are strongly interconnected, while pathways regulating survival and size are not. Interestingly, hESCs/hiPSCs regulate their size by growing exact additional sizes during each cell cycle. Chemical and genetic perturbation studies show that this “adder-model” is dependent on the DNA-damage pathway. In the future, the DRUGPATH-approach may help to predict novel pathway interactomes from high-content drug screens.
Collapse
Affiliation(s)
- Boris Kovacic
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna (MUV), Vienna, Austria.
| | - Margit Rosner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna (MUV), Vienna, Austria
| | - Karin Schlangen
- Section for Biosimulation and Bioinformatics, Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna (MUV), Vienna, Austria
| | - Nina Kramer
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna (MUV), Vienna, Austria
| | - Markus Hengstschläger
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna (MUV), Vienna, Austria
| |
Collapse
|
34
|
Santoro R, Perrucci GL, Gowran A, Pompilio G. Unchain My Heart: Integrins at the Basis of iPSC Cardiomyocyte Differentiation. Stem Cells Int 2019; 2019:8203950. [PMID: 30906328 PMCID: PMC6393933 DOI: 10.1155/2019/8203950] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/20/2018] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
The cellular response to the extracellular matrix (ECM) microenvironment mediated by integrin adhesion is of fundamental importance, in both developmental and pathological processes. In particular, mechanotransduction is of growing importance in groundbreaking cellular models such as induced pluripotent stem cells (iPSC), since this process may strongly influence cell fate and, thus, augment the precision of differentiation into specific cell types, e.g., cardiomyocytes. The decryption of the cellular machinery starting from ECM sensing to iPSC differentiation calls for new in vitro methods. Conveniently, engineered biomaterials activating controlled integrin-mediated responses through chemical, physical, and geometrical designs are key to resolving this issue and could foster clinical translation of optimized iPSC-based technology. This review introduces the main integrin-dependent mechanisms and signalling pathways involved in mechanotransduction. Special consideration is given to the integrin-iPSC linkage signalling chain in the cardiovascular field, focusing on biomaterial-based in vitro models to evaluate the relevance of this process in iPSC differentiation into cardiomyocytes.
Collapse
Affiliation(s)
- Rosaria Santoro
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Gianluca Lorenzo Perrucci
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Aoife Gowran
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Giulio Pompilio
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, via Festa del Perdono 7, Milan, Italy
| |
Collapse
|
35
|
Wei W, Huo B, Shi X. miR-600 inhibits lung cancer via downregulating the expression of METTL3. Cancer Manag Res 2019; 11:1177-1187. [PMID: 30774445 PMCID: PMC6362936 DOI: 10.2147/cmar.s181058] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Methyltransferase like 3 (METTL3) is an RNA methyltransferase implicated in mRNA biogenesis, decay, and translation control through N6-methyladenosine (m6A) modification. Methods To find new treatment strategies for lung cancer and to elucidate the mechanism underlying the phenomenon, we treated the human lung cancer cell lines A549 and H1299 to investigate the effect of METTL3 on lung cancer. Results We observed that knockdown of METTL3 inhibited the survival and proliferation of A549 and H1299 cells. The migration and proliferation of both cell lines were significantly decreased, and the apoptosis was induced in comparison with control cells. These results were further confirmed by the transfection of miRNA of METTL3 increased the Bax/Bcl-2 ratio in A549 and H1299 cells, which is a sign that mitochondrial apoptotic pathway was triggered. The PI3K/Akt pathway is implicated in cell growth and survival and we also observed that knockdown of METTL3 changed the expression and phosphorylation of proteins of PI3K signaling pathway members. Further, our results demonstrated that miR-600 inhibited the expression of METTL3 and reversed the positive effect of METTL3 on NSCLC progression, indicating an miR-600/METTL3 pathway in NSCLC. Conclusion These data suggested that miR-600 inhibited lung cancer via down-regulating METTL3 expression, and knockdown of METTL3 might be used as a novel strategy for lung cancer therapy.
Collapse
Affiliation(s)
- Wenwen Wei
- Department of Respiratory Medicine, The Second People's Hospital of Dongying, Guangrao City, Shandong Province 257335, People's Republic of China,
| | - Baosheng Huo
- Department of Thoracic Surgery, The Second People's Hospital of Dongying, Guangrao City, Shandong Province 257335, People's Republic of China,
| | - Xiulan Shi
- Department of Respiratory Medicine, The Second People's Hospital of Dongying, Guangrao City, Shandong Province 257335, People's Republic of China,
| |
Collapse
|
36
|
Wang X, Liu X, Zhao Y, Sun H, Wang Y. Cytoprotective role of S14G-humanin (HNG) in ultraviolet-B induced epidermal stem cells injury. Biomed Pharmacother 2019; 110:248-253. [DOI: 10.1016/j.biopha.2018.11.059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 10/31/2018] [Accepted: 11/14/2018] [Indexed: 12/24/2022] Open
|
37
|
Yamazaki T, Liu L, Lazarev D, Al-Zain A, Fomin V, Yeung PL, Chambers SM, Lu CW, Studer L, Manley JL. TCF3 alternative splicing controlled by hnRNP H/F regulates E-cadherin expression and hESC pluripotency. Genes Dev 2018; 32:1161-1174. [PMID: 30115631 PMCID: PMC6120717 DOI: 10.1101/gad.316984.118] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 06/22/2018] [Indexed: 12/13/2022]
Abstract
Yamazaki et al. show that alternative splicing creates two TCF3 isoforms (E12 and E47) and identified two related splicing factors, hnRNPs H1 and F (hnRNP H/F), that regulate TCF3 splicing. Expression of known TCF3 target E-cadherin, critical for maintaining ESC pluripotency, is repressed by E47 but not by E12. Alternative splicing (AS) plays important roles in embryonic stem cell (ESC) differentiation. In this study, we first identified transcripts that display specific AS patterns in pluripotent human ESCs (hESCs) relative to differentiated cells. One of these encodes T-cell factor 3 (TCF3), a transcription factor that plays important roles in ESC differentiation. AS creates two TCF3 isoforms, E12 and E47, and we identified two related splicing factors, heterogeneous nuclear ribonucleoproteins (hnRNPs) H1 and F (hnRNP H/F), that regulate TCF3 splicing. We found that hnRNP H/F levels are high in hESCs, leading to high E12 expression, but decrease during differentiation, switching splicing to produce elevated E47 levels. Importantly, hnRNP H/F knockdown not only recapitulated the switch in TCF3 AS but also destabilized hESC colonies and induced differentiation. Providing an explanation for this, we show that expression of known TCF3 target E-cadherin, critical for maintaining ESC pluripotency, is repressed by E47 but not by E12.
Collapse
Affiliation(s)
- Takashi Yamazaki
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Lizhi Liu
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Denis Lazarev
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Amr Al-Zain
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Vitalay Fomin
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Percy Luk Yeung
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Child Health Institute of New Jersey, New Brunswick, New Jersey 08901, USA
| | - Stuart M Chambers
- The Center for Stem Cell Biology, Sloan Kettering Institute, New York, New York 10065, USA.,Developmental Biology Program, Sloan Kettering Institute, New York, New York 10065, USA
| | - Chi-Wei Lu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Child Health Institute of New Jersey, New Brunswick, New Jersey 08901, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan Kettering Institute, New York, New York 10065, USA.,Developmental Biology Program, Sloan Kettering Institute, New York, New York 10065, USA
| | - James L Manley
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| |
Collapse
|
38
|
Park J, Son Y, Lee NG, Lee K, Lee DG, Song J, Lee J, Kim S, Cho MJ, Jang JH, Lee J, Park JG, Kim YG, Kim JS, Lee J, Cho YS, Park YJ, Han BS, Bae KH, Han S, Kang B, Haam S, Lee SH, Lee SC, Min JK. DSG2 Is a Functional Cell Surface Marker for Identification and Isolation of Human Pluripotent Stem Cells. Stem Cell Reports 2018; 11:115-127. [PMID: 29910125 PMCID: PMC6117473 DOI: 10.1016/j.stemcr.2018.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 05/17/2018] [Accepted: 05/17/2018] [Indexed: 11/28/2022] Open
Abstract
Pluripotent stem cells (PSCs) represent the most promising clinical source for regenerative medicine. However, given the cellular heterogeneity within cultivation and safety concerns, the development of specific and efficient tools to isolate a pure population and eliminate all residual undifferentiated PSCs from differentiated derivatives is a prerequisite for clinical applications. In this study, we raised a monoclonal antibody and identified its target antigen as desmoglein-2 (DSG2). DSG2 co-localized with human PSC (hPSC)-specific cell surface markers, and its expression was rapidly downregulated upon differentiation. The depletion of DSG2 markedly decreased hPSC proliferation and pluripotency marker expression. In addition, DSG2-negative population in hPSCs exhibited a notable suppression in embryonic body and teratoma formation. The actions of DSG2 in regulating the self-renewal and pluripotency of hPSCs were predominantly exerted through the regulation of β-catenin/Slug-mediated epithelial-to-mesenchymal transition. Our results demonstrate that DSG2 is a valuable PSC surface marker that is essential for the maintenance of PSC self-renewal. DSG2 is a valuable cell surface marker for defining the state of pluripotency in PSCs DSG2 is essential for the maintenance of PSC self-renewal and pluripotency DSG2 regulates β-catenin-mediated EMT signaling in PSCs DSG2 is essential for the acquisition of pluripotency during reprogramming
Collapse
Affiliation(s)
- Jongjin Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Yeonsung Son
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Na Geum Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Kyungmin Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Dong Gwang Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Jinhoi Song
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Jaemin Lee
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Seokho Kim
- Aging Research Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Min Ji Cho
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Ju-Hong Jang
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Jangwook Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Jong-Gil Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Yeon-Gu Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Jang-Seong Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Jungwoon Lee
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Yee Sook Cho
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Young-Jun Park
- Research Center for Metabolic Regulation, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Baek Soo Han
- Research Center for Metabolic Regulation, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Kwang-Hee Bae
- Research Center for Metabolic Regulation, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Seungmin Han
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 03722, Korea
| | - Byunghoon Kang
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 03722, Korea
| | - Seungjoo Haam
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul 03722, Korea
| | - Sang-Hyun Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea.
| | - Sang Chul Lee
- Research Center for Metabolic Regulation, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea.
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34141, Korea.
| |
Collapse
|
39
|
Laco F, Woo TL, Zhong Q, Szmyd R, Ting S, Khan FJ, Chai CLL, Reuveny S, Chen A, Oh S. Unraveling the Inconsistencies of Cardiac Differentiation Efficiency Induced by the GSK3β Inhibitor CHIR99021 in Human Pluripotent Stem Cells. Stem Cell Reports 2018; 10:1851-1866. [PMID: 29706502 PMCID: PMC5989659 DOI: 10.1016/j.stemcr.2018.03.023] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 03/28/2018] [Accepted: 03/28/2018] [Indexed: 12/21/2022] Open
Abstract
Cardiac differentiation efficiency is hampered by inconsistencies and low reproducibility. We analyzed the differentiation process of multiple human pluripotent stem cell (hPSC) lines in response to dynamic GSK3β inhibition under varying cell culture conditions. hPSCs showed strong differences in cell-cycle profiles with varying culture confluency. hPSCs with a higher percentage of cells in the G1 phase of the cell cycle exhibited cell death and required lower doses of GSK3β inhibitors to induce cardiac differentiation. GSK3β inhibition initiated cell-cycle progression via cyclin D1 and modulated both Wnt signaling and the transcription factor (TCF) levels, resulting in accelerated or delayed mesoderm differentiation. The TCF levels were key regulators during hPSC differentiation with CHIR99021. Our results explain how differences in hPSC lines and culture conditions impact cell death and cardiac differentiation. By analyzing the cell cycle, we were able to select for highly cardiogenic hPSC lines and increase the experimental reproducibility by predicting differentiation outcomes. Lineage variety and cell culture density affect the cell cycle in hPSCs CHIR99021 is cytotoxic to hPSCs with reduced S/G2/M cell-cycle phases Cardiac differentiation reproducibility depends on cell-cycle consistency in hPSCs Cell cycle and TCF protein levels modulate CHIR99021-induced differentiation
Collapse
Affiliation(s)
- Filip Laco
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore.
| | - Tsung Liang Woo
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Qixing Zhong
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Radoslaw Szmyd
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos #03-01, Singapore 138673, Singapore
| | - Sherwin Ting
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Fahima Jaleel Khan
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Christina L L Chai
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Shaul Reuveny
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Allen Chen
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Steve Oh
- Bioprocessing Technology Institute, 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore.
| |
Collapse
|
40
|
Sulaiman A, McGarry S, Li L, Jia D, Ooi S, Addison C, Dimitroulakos J, Arnaout A, Nessim C, Yao Z, Ji G, Song H, Gadde S, Li X, Wang L. Dual inhibition of Wnt and Yes-associated protein signaling retards the growth of triple-negative breast cancer in both mesenchymal and epithelial states. Mol Oncol 2018; 12:423-440. [PMID: 29316250 PMCID: PMC5891054 DOI: 10.1002/1878-0261.12167] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/24/2017] [Accepted: 12/01/2017] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancer (TNBC), the most refractory subtype of breast cancer to current treatments, accounts disproportionately for the majority of breast cancer-related deaths. This is largely due to cancer plasticity and the development of cancer stem cells (CSCs). Recently, distinct yet interconvertible mesenchymal-like and epithelial-like states have been revealed in breast CSCs. Thus, strategies capable of simultaneously inhibiting bulk and CSC populations in both mesenchymal and epithelial states have yet to be developed. Wnt/β-catenin and Hippo/YAP pathways are crucial in tumorigenesis, but importantly also possess tumor suppressor functions in certain contexts. One possibility is that TNBC cells in epithelial or mesenchymal state may differently affect Wnt/β-catenin and Hippo/YAP signaling and CSC phenotypes. In this report, we found that YAP signaling and CD44high /CD24-/low CSCs were upregulated while Wnt/β-catenin signaling and ALDH+ CSCs were downregulated in mesenchymal-like TNBC cells, and vice versa in their epithelial-like counterparts. Dual knockdown of YAP and Wnt/β-catenin, but neither alone, was required for effective suppression of both CD44high /CD24-/low and ALDH+ CSC populations in mesenchymal and epithelial TNBC cells. These observations were confirmed with cultured tumor fragments prepared from patients with TNBC after treatment with Wnt inhibitor ICG-001 and YAP inhibitor simvastatin. In addition, a clinical database showed that decreased gene expression of Wnt and YAP was positively correlated with decreased ALDH and CD44 expression in patients' samples while increased patient survival. Furthermore, tumor growth of TNBC cells in either epithelial or mesenchymal state was retarded, and both CD44high /CD24-/low and ALDH+ CSC subpopulations were diminished in a human xenograft model after dual administration of ICG-001 and simvastatin. Tumorigenicity was also hampered after secondary transplantation. These data suggest a new therapeutic strategy for TNBC via dual Wnt and YAP inhibition.
Collapse
Affiliation(s)
- Andrew Sulaiman
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaCanada
- China‐Canada Centre of Research for Digestive DiseasesUniversity of OttawaCanada
- Institute of Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineChina
- Ottawa Institute of Systems BiologyUniversity of OttawaCanada
| | - Sarah McGarry
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaCanada
| | - Li Li
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaCanada
- China‐Canada Centre of Research for Digestive DiseasesUniversity of OttawaCanada
- Institute of Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineChina
- Ottawa Institute of Systems BiologyUniversity of OttawaCanada
| | - Deyong Jia
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaCanada
- China‐Canada Centre of Research for Digestive DiseasesUniversity of OttawaCanada
- Institute of Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineChina
- Ottawa Institute of Systems BiologyUniversity of OttawaCanada
| | - Sarah Ooi
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaCanada
| | - Christina Addison
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaCanada
- Centre for Cancer TherapeuticsOttawa Hospital Research InstituteCanada
| | - Jim Dimitroulakos
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaCanada
- Centre for Cancer TherapeuticsOttawa Hospital Research InstituteCanada
| | - Angel Arnaout
- Centre for Cancer TherapeuticsOttawa Hospital Research InstituteCanada
| | - Carolyn Nessim
- Centre for Cancer TherapeuticsOttawa Hospital Research InstituteCanada
| | - Zemin Yao
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaCanada
- China‐Canada Centre of Research for Digestive DiseasesUniversity of OttawaCanada
- Institute of Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineChina
- Ottawa Institute of Systems BiologyUniversity of OttawaCanada
| | - Guang Ji
- China‐Canada Centre of Research for Digestive DiseasesUniversity of OttawaCanada
- Institute of Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineChina
| | - Haiyan Song
- China‐Canada Centre of Research for Digestive DiseasesUniversity of OttawaCanada
- Institute of Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineChina
| | - Suresh Gadde
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaCanada
| | - Xuguang Li
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaCanada
- Centre for Biologics Evaluation, Biologics and Genetic Therapies DirectorateHealth CanadaSir Frederick G. Banting Research CentreOttawaCanada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and ImmunologyFaculty of MedicineUniversity of OttawaCanada
- China‐Canada Centre of Research for Digestive DiseasesUniversity of OttawaCanada
- Institute of Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineChina
- Ottawa Institute of Systems BiologyUniversity of OttawaCanada
- Regenerative Medicine ProgramOttawa Hospital Research InstituteCanada
| |
Collapse
|
41
|
Abstract
Background Ovarian cancer is one of the most fatal gynecologic malignancies, with most patients diagnosed at the late stage due to insidious onset and lack of early onset specific symptoms. Previous studies have implied that isoliquiritigenin (ILQ) is a promising chemopreventive agent against oral cancer. Aim This study aimed to investigate effects of ILQ and elucidate the related mechanism. Materials and methods Ovarian cancer cell lines, SKOV3 and OVCAR3, were treated with various concentrations of ILQ to detect the dose-dependent effects of ILQ and select the suitable concentration. CCK8 assay and clone formation efficiency assays were used to detect viability and proliferation. The cell migration, invasion, and apoptosis were evaluated by wound healing assays, transwell, and flow cytometry assays. The expression of apoptosis-related proteins (Caspase-3, Caspase3-p17, Bcl-2, Bax, and Bim) and related-signaling pathway proteins were also detected by Western blot. Results It was observed that the treatment of ILQ inhibited the survival and proliferation of SKOV3 and OVCAR3 cells. ILQ treatment inhibited migration and invasion, and induced apoptosis in SKOV3 and OVCAR3 cells. Also, the ILQ treatment increased the Bax/Bcl-2 ratio in SKOV3 and OVCAR3 cells, suggesting that a mitochondrial apoptotic pathway was triggered. It was also observed that, after treated with ILQ, the phosphorylated form of Akt and mTOR decreased and the expression of GSK3β increased, while P70/S6K decreased. ILQ treatment also decreased the expression of Wnt3a and, therefore, caused the decrease of phosphorylated ERK. ILQ also suppressed the PI3K/Akt/mTOR pathway by reduced the expression level of p-Akt, p-mTOR, P70/S6K and Cyclin D1 in Ishikawa and ES-2 cells. Conclusion The data suggested that ILQ inhibited viability, proliferation, and invasion, and induced apoptosis of SKOV3 and OVCAR3 cells through the PI3K/Akt/mTOR pathway. Together, the data revealed that ILQ treatment may be used as a novel strategy for ovarian cancer therapy.
Collapse
Affiliation(s)
- Nan Li
- Department of Gynecology, Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Liang Yang
- Department of Neurosurgery, Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Xinna Deng
- Department of Oncology & Immunotherapy, Hebei General Hospital, Shijiazhuang, People's Republic of China
| | - Yanan Sun
- Department of Obstetrics and Gynecology, Bethune International Peace Hospital of PLA, Shijiazhuang, People's Republic of China
| |
Collapse
|
42
|
VE-Cadherin regulates the self-renewal of mouse embryonic stem cells via LIF/Stat3 signaling pathway. Biomaterials 2018; 158:34-43. [DOI: 10.1016/j.biomaterials.2017.12.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 12/08/2017] [Accepted: 12/15/2017] [Indexed: 01/01/2023]
|
43
|
Cheng T, Zhai K, Chang Y, Yao G, He J, Wang F, Kong H, Xin H, Wang H, Jin M, Gong B, Gu L, Yang Z, Wu Y, Ji G, Sun Y. CHIR99021 combined with retinoic acid promotes the differentiation of primordial germ cells from human embryonic stem cells. Oncotarget 2018; 8:7814-7826. [PMID: 27999196 PMCID: PMC5352363 DOI: 10.18632/oncotarget.13958] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 12/01/2016] [Indexed: 11/25/2022] Open
Abstract
Primordial germ cells (PGCs) derived from human embryonic stem cells (hESCs) represent as a desirable experimental model as well as a potential strategy for treating male infertility. Here, we developed a simple and feasible method for differentiation of PGCs from hESCs by using CHIR99021 (an inhibitor of glycogen synthase kinase 3) and retinoic acid (RA). We firstly found that the deleted in azoospermia-like (DAZL) protein can be detected in 3 d CHIR99021 plus 9 d retinoic acid treated cultures and 12 d CHIR99021 plus retinoic acid co-treated cultures, but not expressed in single CHIR99021 treated cultures, single retinoic acid treated cultures, as well as 3 d retinoic acid plus 9 d CHIR99021 treated cultures. Next, we showed that several PGCs’ markers were expressed in the 12 d CHIR99021 and retinoic acid co-treated cultures or 3 d CHIR99021 plus 9 d retinoic acid treated cultures. Moreover, meiosis was initiated in CHIR99021 and retinoic acid co-treated cultures as evidenced by a significant expression of the punctate synaptonemal complex protein 3 (SCP3). Fluorescent in situ hybridization (FISH) analysis indicated that a small percentage of putative 1N populations were formed. Mechanically, we found that β-catenin relocated into nucleus after the treatment of 3 d CHIR99021 suggesting that Wnt signaling pathway was activated. Furthermore, blockade of Wnt signaling pathway by IWR-1 can reverse CHIR99021 and retinoic acid mediated-effects. Taken together, our results indicate that CHIR99021 combined with retinoic acid can effectively differentiate hESCs into PGCs via activating Wnt signaling pathway.
Collapse
Affiliation(s)
- Tingting Cheng
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kui Zhai
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yan Chang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Guidong Yao
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiahuan He
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fang Wang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huijuan Kong
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hang Xin
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huiwen Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Meng Jin
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Bing Gong
- Department of Cardiac Surgery, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Gu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhiguang Yang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yanyun Wu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Guangju Ji
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yingpu Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
44
|
Cui N, Yang WT, Zheng PS. Slug inhibits the proliferation and tumor formation of human cervical cancer cells by up-regulating the p21/p27 proteins and down-regulating the activity of the Wnt/β-catenin signaling pathway via the trans-suppression Akt1/p-Akt1 expression. Oncotarget 2018; 7:26152-67. [PMID: 27036045 PMCID: PMC5041971 DOI: 10.18632/oncotarget.8434] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/14/2016] [Indexed: 11/17/2022] Open
Abstract
Slug (Snai2) has been demonstrated to act as an oncogene or tumor suppressor in different human cancers, but the function of Slug in cervical cancer remains poorly understood. In this study, we demonstrated that Slug could suppress the proliferation of cervical cancer cells in vitro and tumor formation in vivo. Further experiments found that Slug could trans-suppress the expression of Akt1/p-Akt1 by binding to E-box motifs in the promoter of the Akt1 gene and then inhibit the cell proliferation and tumor formation of cervical cancer cells by up-regulating p21/p27 and/or down-regulating the activity of the Wnt/β-catenin signaling pathway. Therefore, Slug acts as a tumor suppressor during cervical carcinogenesis.
Collapse
Affiliation(s)
- Nan Cui
- Department of Reproductive Medicine, First Affiliated Hospital, Xi'an Jiaotong University Medical School, Xi'an, The People's Republic of China.,Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Medical School, Xi'an, The People's Republic of China
| | - Wen-Ting Yang
- Department of Reproductive Medicine, First Affiliated Hospital, Xi'an Jiaotong University Medical School, Xi'an, The People's Republic of China.,Section of Cancer Stem Cell Research, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of the People's Republic of China, Xi'an, The People's Republic of China
| | - Peng-Sheng Zheng
- Department of Reproductive Medicine, First Affiliated Hospital, Xi'an Jiaotong University Medical School, Xi'an, The People's Republic of China.,Section of Cancer Stem Cell Research, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of the People's Republic of China, Xi'an, The People's Republic of China
| |
Collapse
|
45
|
Kim JY, Kim SY, Choi HS, Kim MK, Lee HM, Jang YJ, Ryu CJ. Progesterone Receptor Membrane Component 1 suppresses the p53 and Wnt/β-catenin pathways to promote human pluripotent stem cell self-renewal. Sci Rep 2018; 8:3048. [PMID: 29445107 PMCID: PMC5813096 DOI: 10.1038/s41598-018-21322-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/02/2018] [Indexed: 12/22/2022] Open
Abstract
Progesterone receptor membrane component 1 (PGRMC1) is a multifunctional heme-binding protein involved in various diseases, including cancers and Alzheimer’s disease. Previously, we generated two monoclonal antibodies (MAbs) 108-B6 and 4A68 against surface molecules on human pluripotent stem cells (hPSCs). Here we show that PGRMC1 is the target antigen of both MAbs, and is predominantly expressed on hPSCs and some cancer cells. PGRMC1 is rapidly downregulated during early differentiation of hPSCs. Although PGRMC1 knockdown leads to a spread-out morphology and impaired self-renewal in hPSCs, PGRMC1 knockdown hPSCs do not show apoptosis and autophagy. Instead, PGRMC1 knockdown leads to differentiation of hPSCs into multiple lineage cells without affecting the expression of pluripotency markers. PGRMC1 knockdown increases cyclin D1 expression and decreases Plk1 expression in hPSCs. PGRMC1 knockdown also induces p53 expression and stability, suggesting that PGRMC1 maintains hPSC self-renewal through suppression of p53-dependent pathway. Analysis of signaling molecules further reveals that PGRMC1 knockdown promotes inhibitory phosphorylation of GSK-3β and increased expression of Wnt3a and β-catenin, which leads to activation of Wnt/β-catenin signaling. The results suggest that PGRMC1 suppresses the p53 and Wnt/β-catenin pathways to promote self-renewal and inhibit early differentiation in hPSCs.
Collapse
Affiliation(s)
- Ji Yea Kim
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, Korea
| | - So Young Kim
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, Korea
| | - Hong Seo Choi
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, Korea
| | - Min Kyu Kim
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, Korea
| | - Hyun Min Lee
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, Korea
| | - Young-Joo Jang
- Department of Nanobiomedical Science, BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Korea.
| | - Chun Jeih Ryu
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, Korea.
| |
Collapse
|
46
|
Tian X, Tao F, Zhang B, Dong JT, Zhang Z. The miR-203/SNAI2 axis regulates prostate tumor growth, migration, angiogenesis and stemness potentially by modulating GSK-3β/β-CATENIN signal pathway. IUBMB Life 2018; 70:224-236. [PMID: 29389061 DOI: 10.1002/iub.1720] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/09/2018] [Indexed: 12/16/2022]
Abstract
Dysregulation of microRNA expression plays a pivotal role in the initiation and progression of a variety of human carcinomas including prostate cancer. Our previous studies have demonstrated that the silence of miR-203 contributes to the invasiveness of malignant breast cancer cells by targeting SNAI2. However, the effects and underlying mechanisms of miR-203/SNAI2 axis in prostate cancer have not been elucidated. The aim of this study is to explore the effects of miR-203/SNAI2 axis on the biological characteristics of prostate carcinomas both in vitro and in vivo. We found that miR-203 was significantly downregulated in prostate cancer cell lines compared with immortalized prostate epithelial cells using semi-quantitative PCR and real-time PCR, as well as in clinical prostate cancer tissues compared to normal tissues using TCGA analysis. Functionally, miR-203 inhibited prostate cancer cell proliferation, migration, endothelial cell tube formation and cancer stemness in vitro. Meanwhile, overexpression of miR-203 suppressed SNAI2 expression both in DU145 and PC3 cells. In addition, the in vivo study showed that miR-203 suppressed tumorigenicity, metastasis and angiogenesis of DU145 cells. Ectopic expression of SNAI2 rescued the inhibitory effects of miR-203 both in vitro and in vivo. Importantly, the EMT markers CDH1 and VIMENTIN were modulated by the miR-203/SNAI2 axis. Furthermore, the GSK-3β/β-CATENIN signal pathway was suppressed by miR-203 and could be reactivated by SNAI2. Taken together, this research unveiled the function of miR-203/SNAI2 axis in tumorigenesis, angiogenesis, stemness, metastasis and GSK-3β/β-CATENIN signal pathway in prostate cancer and gave insights into miR-203/SNAI2-targeting therapy for prostate cancer patients. © 2018 IUBMB Life, 70(3):224-236, 2018.
Collapse
Affiliation(s)
- Xinxin Tian
- Tianjin International Joint Academy of Biomedicine (TJAB), Tianjin, People's Republic of China.,Department of Biochemistry and Biophysics, Texas A&M University and Texas AgriLife Research, College Station, TX, USA
| | - Fangfang Tao
- Department of Immunology and Microbiology, Basic Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Baotong Zhang
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Emory Winship Cancer Institute, Atlanta, GA, USA
| | - Jin-Tang Dong
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Emory Winship Cancer Institute, Atlanta, GA, USA.,Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Zhiqian Zhang
- Tianjin International Joint Academy of Biomedicine (TJAB), Tianjin, People's Republic of China.,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, People's Republic of China
| |
Collapse
|
47
|
Kim MH, Kino-oka M. Bioprocessing Strategies for Pluripotent Stem Cells Based on Waddington’s Epigenetic Landscape. Trends Biotechnol 2018; 36:89-104. [DOI: 10.1016/j.tibtech.2017.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 10/02/2017] [Accepted: 10/10/2017] [Indexed: 12/12/2022]
|
48
|
Lin SY, Lee YX, Yu SL, Chang GC, Chen JJW. Phosphatase of regenerating liver-3 inhibits invasiveness and proliferation in non-small cell lung cancer by regulating the epithelial-mesenchymal transition. Oncotarget 2017; 7:21799-811. [PMID: 26967563 PMCID: PMC5008324 DOI: 10.18632/oncotarget.7985] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 02/16/2016] [Indexed: 01/03/2023] Open
Abstract
Phosphatase of regenerating liver-3 (PRL-3) has been reported to be associated with colon and gastric cancer metastasis. However, the role and function of PRL-3 in human non-small cell lung cancer cells is unknown. Our studies showed that the expression of PRL-3mRNA and protein are higher in less invasive human lung adenocarcinoma cells than in highly invasive cell lines. Ectopic expression of PRL-3 reduced cell capacity for anchorage-dependent growth, anchorage-independent growth, migration, and invasion in vitro, as well as tumorigenesis in vivo. Conversely, catalytic (C104S) and prenylation-site (C170S) mutants enhanced cell invasion. Microarray profiling of PRL-3 transfectants revealed the pathways potentially involving PRL-3, including the epithelial-mesenchymal transition (EMT), extracellular matrix remodeling, and the WNT signaling pathway. Furthermore, we demonstrated that increased PRL-3 reduced Slug and enhanced E-cadherin gene expression through the AKT/GSK3β/β-catenin pathway. In conclusion, our data suggest that PRL-3 might play a tumor suppressor role in lung cancer, distinct from other cancers, by inhibiting EMT-related pathways.
Collapse
Affiliation(s)
- Sheng-Yi Lin
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.,Agricultural Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Yue-Xun Lee
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Sung-Liang Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Gee-Chen Chang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.,Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jeremy J W Chen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.,Agricultural Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
49
|
Singh SK, Baker R, Sikkink SK, Nizard C, Schnebert S, Kurfurst R, Tobin DJ. E-cadherin mediates ultraviolet radiation- and calcium-induced melanin transfer in human skin cells. Exp Dermatol 2017. [PMID: 28636748 DOI: 10.1111/exd.13395] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Skin pigmentation is directed by epidermal melanin units, characterized by long-lived and dendritic epidermal melanocytes (MC) that interact with viable keratinocytes (KC) to contribute melanin to the epidermis. Previously, we reported that MC:KC contact is required for melanosome transfer that can be enhanced by filopodi, and by UVR/UVA irradiation, which can upregulate melanosome transfer via Myosin X-mediated control of MC filopodia. Both MC and KC express Ca2+ -dependent E-cadherins. These homophilic adhesion contacts induce transient increases in intra-KC Ca2+ , while ultraviolet radiation (UVR) raises intra-MC Ca2+ via calcium-selective ORAI1 ion channels; both are associated with regulating melanogenesis. However, how Ca2+ triggers melanin transfer remains unclear. Here we evaluated the role of E-cadherin in UVR-mediated melanin transfer in human skin cells. MC and KC in human epidermis variably express filopodia-associated E-cadherin, Cdc42, VASP and β-catenin, all of which were upregulated by UVR in human MC in vitro. Knockdown of E-cadherin revealed that this cadherin is essential for UVR-induced MC filopodia formation and melanin transfer. Moreover, Ca2+ induced a dose-dependent increase in filopodia formation and melanin transfer, as well as increased β-catenin, Cdc42, Myosin X and E-cadherin expression in these skin cells. Together, these data suggest that filopodial proteins and E-cadherin, which are upregulated by intracellular (UVR-stimulated) and extracellular Ca2+ availability, are required for filopodia formation and melanin transfer. This may open new avenues to explore how Ca2+ signalling influences human pigmentation.
Collapse
Affiliation(s)
- Suman K Singh
- Centre for Skin Sciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| | - Richard Baker
- Centre for Skin Sciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| | - Stephen K Sikkink
- Centre for Skin Sciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| | | | | | | | - Desmond J Tobin
- Centre for Skin Sciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
50
|
Zhao H, Jin Y. Signaling networks in the control of pluripotency. Curr Opin Genet Dev 2017; 46:141-148. [PMID: 28806594 DOI: 10.1016/j.gde.2017.07.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/17/2017] [Accepted: 07/27/2017] [Indexed: 11/16/2022]
Abstract
Embryonic stem cells (ESCs) are characterized by their ability of unlimited self-renewal in vitro and pluripotent developmental potential, which endows them with great values in basic research and future clinical application. However, realization of full potential of ESCs is dependent on the elucidation of molecular mechanisms governing ESCs, among which signaling pathways play critical roles. A great deal of efforts has been made in the past decades to understand what and how signaling pathways contribute to the establishment and maintenance of pluripotency. In this review, we discuss signaling networks in both mouse and human ESCs, focusing on signals involved in the control of self-renewal and differentiation. In addition, the modulation of signaling pathways by pluripotency-associated transcription factors is also briefly summarized.
Collapse
Affiliation(s)
- Hanzhi Zhao
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Ying Jin
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai, China.
| |
Collapse
|