51
|
Kumar V, Park S, Lee U, Kim J. The Organizer and Its Signaling in Embryonic Development. J Dev Biol 2021; 9:jdb9040047. [PMID: 34842722 PMCID: PMC8628936 DOI: 10.3390/jdb9040047] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/20/2021] [Accepted: 10/29/2021] [Indexed: 12/25/2022] Open
Abstract
Germ layer specification and axis formation are crucial events in embryonic development. The Spemann organizer regulates the early developmental processes by multiple regulatory mechanisms. This review focuses on the responsive signaling in organizer formation and how the organizer orchestrates the germ layer specification in vertebrates. Accumulated evidence indicates that the organizer influences embryonic development by dual signaling. Two parallel processes, the migration of the organizer’s cells, followed by the transcriptional activation/deactivation of target genes, and the diffusion of secreting molecules, collectively direct the early development. Finally, we take an in-depth look at active signaling that originates from the organizer and involves germ layer specification and patterning.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea;
| | - Soochul Park
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea;
| | - Unjoo Lee
- Department of Electrical Engineering, Hallym University, Chuncheon 24252, Korea
- Correspondence: (U.L.); (J.K.); Tel.: +82-33-248-2544 (J.K.); Fax: +82-33-244-8425 (J.K.)
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea;
- Correspondence: (U.L.); (J.K.); Tel.: +82-33-248-2544 (J.K.); Fax: +82-33-244-8425 (J.K.)
| |
Collapse
|
52
|
RSV Promotes Epithelial Neuroendocrine Phenotype Differentiation through NODAL Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9956078. [PMID: 34541002 PMCID: PMC8445725 DOI: 10.1155/2021/9956078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 08/16/2021] [Indexed: 01/04/2023]
Abstract
Background Respiratory syncytial virus (RSV) infects infants and children, predisposing them to development of asthma during adulthood. Epithelial neuroendocrine phenotypes may be associated with development of asthma. This study hopes to ascertain if RSV infection promotes epithelial neuroendocrine phenotypes through the NODAL signaling pathway. Methods The GSE6802 data set was obtained from the GEO database, and the differential genes were analyzed using the R language. An in vitro model was constructed with RSV infected human respiratory epithelial cells, and then real-time qPCR and immunofluorescence were used to detect the expression of different epithelial biomarkers and airway neuropeptides. The acute and chronic infection model of RSV infection was established by intranasal injection of RSV into guinea pigs. Immunohistochemistry and Western blot were used to detect the expression of pulmonary neuroendocrine cells markers ENO2 and neuropeptides. Results The expression levels of ENO2, SP, CGRP, and NODAL/ACTRII were significantly higher in the RSV infection group than those of the control group, which were abrogated by siRNA-NODAL. In vivo, we found that the expression levels of ENO2, SP, and CGRP were significantly higher than that of the control group. Conclusion RSV promotes epithelial neuroendocrine phenotypes through the NODAL signaling pathway.
Collapse
|
53
|
Anlas K, Trivedi V. Studying evolution of the primary body axis in vivo and in vitro. eLife 2021; 10:e69066. [PMID: 34463611 PMCID: PMC8456739 DOI: 10.7554/elife.69066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/27/2021] [Indexed: 02/06/2023] Open
Abstract
The metazoan body plan is established during early embryogenesis via collective cell rearrangements and evolutionarily conserved gene networks, as part of a process commonly referred to as gastrulation. While substantial progress has been achieved in terms of characterizing the embryonic development of several model organisms, underlying principles of many early patterning processes nevertheless remain enigmatic. Despite the diversity of (pre-)gastrulating embryo and adult body shapes across the animal kingdom, the body axes, which are arguably the most fundamental features, generally remain identical between phyla. Recently there has been a renewed appreciation of ex vivo and in vitro embryo-like systems to model early embryonic patterning events. Here, we briefly review key examples and propose that similarities in morphogenesis and associated gene expression dynamics may reveal an evolutionarily conserved developmental mode as well as provide further insights into the role of external or extraembryonic cues in shaping the early embryo. In summary, we argue that embryo-like systems can be employed to inform previously uncharted aspects of animal body plan evolution as well as associated patterning rules.
Collapse
Affiliation(s)
| | - Vikas Trivedi
- EMBL BarcelonaBarcelonaSpain
- EMBL Heidelberg, Developmental BiologyHeidelbergGermany
| |
Collapse
|
54
|
Bruveris FF, Ng ES, Stanley EG, Elefanty AG. VEGF, FGF2, and BMP4 regulate transitions of mesoderm to endothelium and blood cells in a human model of yolk sac hematopoiesis. Exp Hematol 2021; 103:30-39.e2. [PMID: 34437953 DOI: 10.1016/j.exphem.2021.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022]
Abstract
Exogenous growth factors play an important role in mediating hematopoietic differentiation of human pluripotent stem cells. We explored the role of different factors in early human blood cell production using blast colony formation in methylcellulose as a surrogate assay for yolk sac hematopoiesis. A reporter cell line that read out endothelial (SOX17+) and hematopoietic (RUNX1C+) progenitors facilitated the identification of basic fibroblast growth and vascular endothelial growth factor as critical signals for the progression of mesoderm into endothelium. Bone morphogenetic protein 4 was needed for the subsequent generation of blood from hemogenic endothelium, and this was antagonized by Activin A or high concentrations of the WNT agonist CHIR-99021. Manipulations of the Hedgehog pathway or inhibition of Notch signaling reduced blast colony frequency but did not perturb cell differentiation. These data help to define distinct roles for prerequisite growth factors that commit mesoderm to hemogenic endothelium and subsequently allocate cells to blood lineages.
Collapse
Affiliation(s)
- Freya F Bruveris
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth S Ng
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria, Australia
| | - Edouard G Stanley
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Andrew G Elefanty
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, Victoria, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
55
|
Luo WJ, He SW, Zou WQ, Zhao Y, He QM, Yang XJ, Guo R, Mao YP. Epstein-Barr virus microRNA BART10-3p promotes dedifferentiation and proliferation of nasopharyngeal carcinoma by targeting ALK7. Exp Biol Med (Maywood) 2021; 246:2618-2629. [PMID: 34424090 DOI: 10.1177/15353702211037261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Non-keratinizing nasopharyngeal carcinoma, the major subtype of nasopharyngeal carcinoma, is characterized by low differentiation and a close relation to Epstein-Barr virus infection, which indicates a link between Epstein-Barr virus oncogenesis and loss of differentiation, and raises our interest in investigating the involvement of Epstein-Barr virus in nasopharyngeal carcinoma dedifferentiation. Our previous study showed abundant expression of an Epstein-Barr virus-encoded microRNA, BART10-3p, in nasopharyngeal carcinoma tissues, but the association between BART10-3p and nasopharyngeal carcinoma differentiation remains unknown. Here, we examined the expression and prognostic value of BART10-3p, and undertook bioinformatics analysis and functional assays to investigate the influence of BART10-3p on nasopharyngeal carcinoma differentiation and proliferation and the underpinning mechanism. Microarray analysis identified BART10-3p as the most significantly upregulated Epstein-Barr virus-encoded microRNA in nasopharyngeal carcinoma tissues and the upregulation was confirmed in two public datasets. The expression of BART10-3p was an independent unfavorable prognosticator in nasopharyngeal carcinoma and its integration with the clinical stage showed improved prognosis predictive performance. Bioinformatics analysis suggested a potential role of BART10-3p in tumor differentiation and progression. Functional assays demonstrated that BART10-3p could promote nasopharyngeal carcinoma cell dedifferentiation, epithelial-mesenchymal transition, and proliferation in vitro, and tumorigenicity in vivo. Mechanistically, BART10-3p directly targeted the 3'UTR of ALK7 and suppressed its expression. Reconstitution of ALK7 rescued BART10-3p-induced malignant phenotypes. Overall, our study demonstrates that BART10-3p promotes dedifferentiation and proliferation of nasopharyngeal carcinoma by targeting ALK7, suggesting a promising therapeutic opportunity to reverse the malignant phenotypes of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Wei-Jie Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shi-Wei He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Wen-Qing Zou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yin Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Qing-Mei He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xiao-Jing Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Rui Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yan-Ping Mao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
56
|
Pasqua M, Di Gesù R, Chinnici CM, Conaldi PG, Francipane MG. Generation of Hepatobiliary Cell Lineages from Human Induced Pluripotent Stem Cells: Applications in Disease Modeling and Drug Screening. Int J Mol Sci 2021; 22:8227. [PMID: 34360991 PMCID: PMC8348238 DOI: 10.3390/ijms22158227] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
The possibility to reproduce key tissue functions in vitro from induced pluripotent stem cells (iPSCs) is offering an incredible opportunity to gain better insight into biological mechanisms underlying development and disease, and a tool for the rapid screening of drug candidates. This review attempts to summarize recent strategies for specification of iPSCs towards hepatobiliary lineages -hepatocytes and cholangiocytes-and their use as platforms for disease modeling and drug testing. The application of different tissue-engineering methods to promote accurate and reliable readouts is discussed. Space is given to open questions, including to what extent these novel systems can be informative. Potential pathways for improvement are finally suggested.
Collapse
Affiliation(s)
- Mattia Pasqua
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
| | - Roberto Di Gesù
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
| | - Cinzia Maria Chinnici
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
- Dipartimento della Ricerca, IRCCS ISMETT, 90127 Palermo, Italy;
| | | | - Maria Giovanna Francipane
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
57
|
Arboretto P, Cillo M, Leonardi A. New Insights into Cancer Targeted Therapy: Nodal and Cripto-1 as Attractive Candidates. Int J Mol Sci 2021; 22:ijms22157838. [PMID: 34360603 PMCID: PMC8345935 DOI: 10.3390/ijms22157838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
The transforming growth factor beta (TGF-β) signaling is fundamental for correct embryonic development. However, alterations of this pathway have been correlated with oncogenesis, tumor progression and sustaining of cancer stem cells (CSCs). Cripto-1 (CR-1) and Nodal are two embryonic proteins involved in TGF-β signaling. Their expression is almost undetectable in terminally differentiated cells, but they are often re-expressed in tumor cells, especially in CSCs. Moreover, cancer cells that show high levels of CR-1 and/or Nodal display more aggressive phenotypes in vitro, while in vivo their expression correlates with a worse prognosis in several human cancers. The ability to target CSCs still represents an unmet medical need for the complete eradication of certain types of tumors. Given the prognostic role and the selective expression of CR-1 and Nodal on cancer cells, they represent archetypes for targeted therapy. The aim of this review is to clarify the role of CR-1 and Nodal in cancer stem populations and to summarize the current therapeutic strategy to target CSCs using monoclonal antibodies (mAbs) or other molecular tools to interfere with these two proteins.
Collapse
|
58
|
Hasanpour S, Eagderi S, Poorbagher H, Angrand PO, Hasanpour M, Lashkarbolok M. The effect of Activin pathway modulation on the expression of both pluripotency and differentiation markers during early zebrafish development compared with other vertebrates. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2021; 336:562-575. [PMID: 34254429 DOI: 10.1002/jez.b.23070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 05/22/2021] [Accepted: 06/24/2021] [Indexed: 11/09/2022]
Abstract
Activin-like factors control many developmental processes, including pluripotency maintenance and differentiation. Although Activin-like factors' action in mesendoderm induction has been demonstrated in zebrafish, their involvement in preserving the stemness remains unknown. To investigate the role of maternal Activin-like factors, their effects were promoted or blocked using synthetic human Activin A or SB-431542 treatments respectively until the maternal to zygotic transition. To study the role of zygotic Activin-like factors, SB-431542 treatment was also applied after the maternal to zygotic transition. The effect of the pharmacological modulations of the Activin/Smad pathway was then studied on the mRNA expressions of the ndr1, ndr2, tbxta (no tail/ntl) as the differentiation index, mych, nanog, and oct4 (pou5f3) as the pluripotency markers of the zebrafish embryonic cells as well as sox17 as a definitive endoderm marker. Expression of the target genes was measured at the 16-cell, 256-cell, 1K-cell, oblong, dome, and shield stages using the real-time quantitative polymerase chain reaction (RT-qPCR). Activation of the maternal Activin signaling pathway led to an increase in zygotic expression of the tbxta, particularly marked at the oblong stage. In other words, promotion of the maternal Activin/Smad pathway induced differentiation by advancing the major peaks of ndr1 and nanog, thereby eliciting tbxta expression. Whereas suppression of the maternal or zygotic Activin/Smad pathway sustained the pluripotency by preventing the major peaks of ndr1 and nanog as well as tbxta encoding.
Collapse
Affiliation(s)
- Shaghayegh Hasanpour
- Department of Fisheries, Faculty of Natural Resources, University of Tehran, Karaj, Iran.,Development and Biosystematic Lab., Department of Fisheries and Animal Sciences, Faculty of Natural Resources, University of Tehran, Karaj, Iran
| | - Soheil Eagderi
- Department of Fisheries, Faculty of Natural Resources, University of Tehran, Karaj, Iran
| | - Hadi Poorbagher
- Department of Fisheries, Faculty of Natural Resources, University of Tehran, Karaj, Iran
| | - Pierre-Olivier Angrand
- Univ Lille, CNRS UMR 9020, Inserm UMR-S 1277, CHU Lille, Centre Oscar Lambret, UMR Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Mohammad Hasanpour
- Department of Neurosurgery, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Lashkarbolok
- Department of Radiology, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
59
|
Li B, Qiao C, Jin X, Chan HM. Characterizing the Low-Dose Effects of Methylmercury on the Early Stages of Embryo Development Using Cultured Human Embryonic Stem Cells. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:77007. [PMID: 34328791 PMCID: PMC8323991 DOI: 10.1289/ehp7349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/18/2021] [Accepted: 07/02/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Global concerns of methylmercury (MeHg) exposure have been raised, especially on its effects on pregnant women. Recent epidemiological studies have revealed associations between maternal blood/hair MeHg concentrations, adverse pregnancy outcomes, and developmental deficits. However, the underlying mechanisms remain unclear. OBJECTIVES In this study, we characterized the effects of MeHg exposure on undifferentiated human embryonic stem cells (hESCs) and extrapolated the effects to human embryonic development. METHODS hESCs were exposed to 0, 1, 5, 10, 50, 100 or 200nM MeHg for 24 h or 6 d. Cell adherence and colony formation and expansion were examined under the microscope. Cell attachment, viability/proliferation, apoptosis, stress response, cell cycle, autophagy, and expression of cell lineage marker genes and proteins were measured at the end of exposures. RESULTS Our results indicated that exposure to nanomolar concentrations of MeHg was associated with a) higher levels of reactive oxygen species (ROS) and hemeoxygenase-1 (HO-1), suggesting increased stress and adaptive responses; b) lower cellular adhesion, viability/proliferation, and colony formation and expansion; c) higher levels of apoptosis, reflected by higher cleaved caspase-3 expression and Annexin V binding; d) higher levels of cytoskeleton protein α-tubulin expression; e) higher rates of G1/S phase cell cycle arrest; and f) autophagy inhibition, as shown by a lower LC3BII/LC3BI ratio and accumulation of SQSTM1 (p62). These outcomes were accompanied by higher expressions of self-renewal genes or proteins or both, including OCT4, SOX2, NANOG, and cytokine receptor IL6ST, as well as pluripotency and the cell fate regulator cyclin D1. DISCUSSION These results revealed that under the selection pressure of exposure to low doses of MeHg, some hESCs underwent apoptosis, whereas others adapted and survived with enhanced self-renewal gene expression and specific morphological phenotypes. Findings from the present study provide in vitro evidence that low doses of MeHg adversely affect hESCs when exposed during a period of time that models embryonic pre-, during, and early postimplantation stages. https://doi.org/10.1289/EHP7349.
Collapse
Affiliation(s)
- Bai Li
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Cunye Qiao
- Biostatistics and Modeling Division, Bureau of Food Surveillance and Science Integration, Food Directorate, Health Products and Food Branch (HPFB), Health Canada, Ottawa, Ontario, Canada
| | - Xiaolei Jin
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Food Directorate, HPFB, Health Canada, Ottawa, Ontario, Canada
| | - Hing Man Chan
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
60
|
Madeddu C, Neri M, Sanna E, Oppi S, Macciò A. Experimental Drugs for Chemotherapy- and Cancer-Related Anemia. J Exp Pharmacol 2021; 13:593-611. [PMID: 34194245 PMCID: PMC8238072 DOI: 10.2147/jep.s262349] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/21/2021] [Indexed: 01/03/2023] Open
Abstract
Anemia in cancer patients is a relevant condition complicating the course of the neoplastic disease. Overall, we distinguish the anemia which arises under chemotherapy as pure adverse event of the toxic effects of the drugs used, and the anemia induced by the tumour-associated inflammation, oxidative stress, and systemic metabolic changes, which can be worsened by the concomitant anticancer treatments. This more properly cancer-related anemia depends on several overlapping mechanism, including impaired erythropoiesis and functional iron deficiency, which make its treatment more difficult. Standard therapies approved and recommended for cancer anemia, as erythropoiesis-stimulating agents and intravenous iron administration, are limited to the treatment of chemotherapy-induced anemia, preferably in patients with advanced disease, in view of the still unclear effect of erythropoiesis-stimulating agents on tumour progression and survival. Outside the use of chemotherapy, there are no recommendations for the treatment of cancer-related anemia. For a more complete approach, it is fundamentally a careful evaluation of the type of anemia and iron homeostasis, markers of inflammation and changes in energy metabolism. In this way, anemia management in cancer patient would permit a tailored approach that could give major benefits. Experimental drugs targeting hepcidin and activin II receptor pathways are raising great expectations, and future clinical trials will confirm their role as remedies for cancer-related anemia. Recent evidence on the effect of integrated managements, including nutritional support, antioxidants and anti-inflammatory substances, for the treatment of cancer anemia are emerging. In this review article, we show standard, innovative, and experimental treatment used as remedy for anemia in cancer patients.
Collapse
Affiliation(s)
- Clelia Madeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Manuela Neri
- Department of Gynecologic Oncology, A. Businco Hospital, ARNAS G. Brotzu, Cagliari, Italy
| | - Elisabetta Sanna
- Department of Gynecologic Oncology, A. Businco Hospital, ARNAS G. Brotzu, Cagliari, Italy
| | - Sara Oppi
- Hematology and Transplant Center, A. Businco Hospital, ARNAS G. Brotzu, Cagliari, Italy
| | - Antonio Macciò
- Department of Gynecologic Oncology, A. Businco Hospital, ARNAS G. Brotzu, Cagliari, Italy
| |
Collapse
|
61
|
Liu X, Wang H, Zhao X, Luo Q, Wang Q, Tan K, Wang Z, Jiang J, Cui J, Du E, Xia L, Du W, Chen D, Xia L, Xiao S. Arginine methylation of METTL14 promotes RNA N 6-methyladenosine modification and endoderm differentiation of mouse embryonic stem cells. Nat Commun 2021; 12:3780. [PMID: 34145242 PMCID: PMC8213825 DOI: 10.1038/s41467-021-24035-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/28/2021] [Indexed: 11/08/2022] Open
Abstract
RNA N6-methyladenosine (m6A), the most abundant internal modification of mRNAs, plays key roles in human development and health. Post-translational methylation of proteins is often critical for the dynamic regulation of enzymatic activity. However, the role of methylation of the core methyltransferase METTL3/METTL14 in m6A regulation remains elusive. We find by mass spectrometry that METTL14 arginine 255 (R255) is methylated (R255me). Global mRNA m6A levels are greatly decreased in METTL14 R255K mutant mouse embryonic stem cells (mESCs). We further find that R255me greatly enhances the interaction of METTL3/METTL14 with WTAP and promotes the binding of the complex to substrate RNA. We show that protein arginine N-methyltransferases 1 (PRMT1) interacts with and methylates METTL14 at R255, and consistent with this, loss of PRMT1 reduces mRNA m6A modification globally. Lastly, we find that loss of R255me preferentially affects endoderm differentiation in mESCs. Collectively, our findings show that arginine methylation of METTL14 stabilizes the binding of the m6A methyltransferase complex to its substrate RNA, thereby promoting global m6A modification and mESC endoderm differentiation. This work highlights the crosstalk between protein methylation and RNA methylation in gene expression.
Collapse
Affiliation(s)
- Xiaona Liu
- School of Life Sciences, University of Science and Technology of China, Hefei, P.R. China
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hailong Wang
- Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, P.R. China
| | - Xueya Zhao
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Qizhi Luo
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou, P.R. China
| | - Qingwen Wang
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Kaifen Tan
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Zihan Wang
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Jia Jiang
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Jinru Cui
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Enhui Du
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Linjian Xia
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Wenyi Du
- Sichuan MoDe Technology Co., Ltd, Chengdu, P. R. China
| | - Dahua Chen
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute of Biomedical Research, Yunnan University, Kunming, P.R. China.
| | - Laixin Xia
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China.
- State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, P.R. China.
| | - Shan Xiao
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P.R. China.
| |
Collapse
|
62
|
Lyadova I, Gerasimova T, Nenasheva T. Macrophages Derived From Human Induced Pluripotent Stem Cells: The Diversity of Protocols, Future Prospects, and Outstanding Questions. Front Cell Dev Biol 2021; 9:640703. [PMID: 34150747 PMCID: PMC8207294 DOI: 10.3389/fcell.2021.640703] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/25/2021] [Indexed: 12/23/2022] Open
Abstract
Macrophages (Mφ) derived from induced pluripotent stem cells (iMphs) represent a novel and promising model for studying human Mφ function and differentiation and developing new therapeutic strategies based on or oriented at Mφs. iMphs have several advantages over the traditionally used human Mφ models, such as immortalized cell lines and monocyte-derived Mφs. The advantages include the possibility of obtaining genetically identical and editable cells in a potentially scalable way. Various applications of iMphs are being developed, and their number is rapidly growing. However, the protocols of iMph differentiation that are currently used vary substantially, which may lead to differences in iMph differentiation trajectories and properties. Standardization of the protocols and identification of minimum required conditions that would allow obtaining iMphs in a large-scale, inexpensive, and clinically suitable mode are needed for future iMph applications. As a first step in this direction, the current review discusses the fundamental basis for the generation of human iMphs, performs a detailed analysis of the generalities and the differences between iMph differentiation protocols currently employed, and discusses the prospects of iMph applications.
Collapse
Affiliation(s)
- Irina Lyadova
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | | | | |
Collapse
|
63
|
Soto DA, Navarro M, Zheng C, Halstead MM, Zhou C, Guiltinan C, Wu J, Ross PJ. Simplification of culture conditions and feeder-free expansion of bovine embryonic stem cells. Sci Rep 2021; 11:11045. [PMID: 34040070 PMCID: PMC8155104 DOI: 10.1038/s41598-021-90422-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 05/05/2021] [Indexed: 02/04/2023] Open
Abstract
Bovine embryonic stem cells (bESCs) extend the lifespan of the transient pluripotent bovine inner cell mass in vitro. After years of research, derivation of stable bESCs was only recently reported. Although successful, bESC culture relies on complex culture conditions that require a custom-made base medium and mouse embryonic fibroblasts (MEF) feeders, limiting the widespread use of bESCs. We report here simplified bESC culture conditions based on replacing custom base medium with a commercially available alternative and eliminating the need for MEF feeders by using a chemically-defined substrate. bESC lines were cultured and derived using a base medium consisting of N2B27 supplements and 1% BSA (NBFR-bESCs). Newly derived bESC lines were easy to establish, simple to propagate and stable after long-term culture. These cells expressed pluripotency markers and actively proliferated for more than 35 passages while maintaining normal karyotype and the ability to differentiate into derivatives of all three germ lineages in embryoid bodies and teratomas. In addition, NBFR-bESCs grew for multiple passages in a feeder-free culture system based on vitronectin and Activin A medium supplementation while maintaining pluripotency. Simplified conditions will facilitate the use of bESCs for gene editing applications and pluripotency and lineage commitment studies.
Collapse
Affiliation(s)
- Delia Alba Soto
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA
| | - Micaela Navarro
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA
- Instituto de Investigaciones Biotecnológicas 'Dr Rodolfo Ugalde', UNSAM-CONICET, Buenos Aires, Argentina
| | - Canbin Zheng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | | | - Chuan Zhou
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA
| | - Carly Guiltinan
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA
| | - Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Pablo Juan Ross
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA.
| |
Collapse
|
64
|
Hayes K, Kim YK, Pera MF. A case for revisiting Nodal signaling in human pluripotent stem cells. STEM CELLS (DAYTON, OHIO) 2021; 39:1137-1144. [PMID: 33932319 DOI: 10.1002/stem.3383] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/30/2021] [Indexed: 11/10/2022]
Abstract
Nodal is a transforming growth factor-β (TGF-β) superfamily member that plays a number of critical roles in mammalian embryonic development. Nodal is essential for the support of the peri-implantation epiblast in the mouse embryo and subsequently acts to specify mesendodermal fate at the time of gastrulation and, later, left-right asymmetry. Maintenance of human pluripotent stem cells (hPSCs) in vitro is dependent on Nodal signaling. Because it has proven difficult to prepare a biologically active form of recombinant Nodal protein, Activin or TGFB1 are widely used as surrogates for NODAL in hPSC culture. Nonetheless, the expression of the components of an endogenous Nodal signaling pathway in hPSC provides a potential autocrine pathway for the regulation of self-renewal in this system. Here we review recent studies that have clarified the role of Nodal signaling in pluripotent stem cell populations, highlighted spatial restrictions on Nodal signaling, and shown that Nodal functions in vivo as a heterodimer with GDF3, another TGF-β superfamily member expressed by hPSC. We discuss the role of this pathway in the maintenance of the epiblast and hPSC in light of these new advances.
Collapse
Affiliation(s)
- Kevin Hayes
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | - Yun-Kyo Kim
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | | |
Collapse
|
65
|
Mishra S, Taelman J, Popovic M, Tilleman L, Duthoo E, van der Jeught M, Deforce D, van Nieuwerburgh F, Menten B, de Sutter P, Boel A, Chuva De Sousa Lopes SM, Heindryckx B. Activin A-derived human embryonic stem cells show increased competence to differentiate into primordial germ cell-like cells. Stem Cells 2021; 39:551-563. [PMID: 33470497 PMCID: PMC8248136 DOI: 10.1002/stem.3335] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/21/2020] [Indexed: 12/14/2022]
Abstract
Protocols for specifying human primordial germ cell-like cells (hPGCLCs) from human embryonic stem cells (hESCs) remain hindered by differences between hESC lines, their derivation methods, and maintenance culture conditions. This poses significant challenges for establishing reproducible in vitro models of human gametogenesis. Here, we investigated the influence of activin A (ActA) during derivation and maintenance on the propensity of hESCs to differentiate into PGCLCs. We show that continuous ActA supplementation during hESC derivation (from blastocyst until the formation of the post-inner cell mass intermediate [PICMI]) and supplementation (from the first passage of the PICMI onwards) is beneficial to differentiate hESCs to PGCLCs subsequently. Moreover, comparing isogenic primed and naïve states prior to differentiation, we showed that conversion of hESCs to the 4i-state improves differentiation to (TNAP [tissue nonspecific alkaline phosphatase]+/PDPN [podoplanin]+) PGCLCs. Those PGCLCs expressed several germ cell markers, including TFAP2C (transcription factor AP-2 gamma), SOX17 (SRY-box transcription factor 17), and NANOS3 (nanos C2HC-type zinc finger 3), and markers associated with germ cell migration, CXCR4 (C-X-C motif chemokine receptor 4), LAMA4 (laminin subunit alpha 4), ITGA6 (integrin subunit alpha 6), and CDH4 (cadherin 4), suggesting that the large numbers of PGCLCs obtained may be suitable to differentiate further into more mature germ cells. Finally, hESCs derived in the presence of ActA showed higher competence to differentiate to hPGCLC, in particular if transiently converted to the 4i-state. Our work provides insights into the differences in differentiation propensity of hESCs and delivers an optimized protocol to support efficient human germ cell derivation.
Collapse
Affiliation(s)
- Swati Mishra
- Ghent‐Fertility and Stem cell Team (G‐FAST), Department of Reproductive MedicineGhent University HospitalGhentBelgium
| | - Jasin Taelman
- Ghent‐Fertility and Stem cell Team (G‐FAST), Department of Reproductive MedicineGhent University HospitalGhentBelgium
| | - Mina Popovic
- Ghent‐Fertility and Stem cell Team (G‐FAST), Department of Reproductive MedicineGhent University HospitalGhentBelgium
| | - Laurentijn Tilleman
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical SciencesGhent UniversityGhentBelgium
| | - Evi Duthoo
- Ghent‐Fertility and Stem cell Team (G‐FAST), Department of Reproductive MedicineGhent University HospitalGhentBelgium
| | - Margot van der Jeught
- Ghent‐Fertility and Stem cell Team (G‐FAST), Department of Reproductive MedicineGhent University HospitalGhentBelgium
| | - Dieter Deforce
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical SciencesGhent UniversityGhentBelgium
| | - Filip van Nieuwerburgh
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical SciencesGhent UniversityGhentBelgium
| | - Björn Menten
- Department of Pediatrics and Medical Genetics, Center for Medical GeneticsGhent University HospitalGhentBelgium
| | - Petra de Sutter
- Ghent‐Fertility and Stem cell Team (G‐FAST), Department of Reproductive MedicineGhent University HospitalGhentBelgium
| | - Annekatrien Boel
- Ghent‐Fertility and Stem cell Team (G‐FAST), Department of Reproductive MedicineGhent University HospitalGhentBelgium
| | - Susana M. Chuva De Sousa Lopes
- Ghent‐Fertility and Stem cell Team (G‐FAST), Department of Reproductive MedicineGhent University HospitalGhentBelgium
- Department of Anatomy and EmbryologyLeiden University Medical CentreLeidenThe Netherlands
| | - Björn Heindryckx
- Ghent‐Fertility and Stem cell Team (G‐FAST), Department of Reproductive MedicineGhent University HospitalGhentBelgium
| |
Collapse
|
66
|
Webster NJ, Maywald RL, Benton SM, Dawson EP, Murillo OD, LaPlante EL, Milosavljevic A, Lanza DG, Heaney JD. Testicular germ cell tumors arise in the absence of sex-specific differentiation. Development 2021; 148:260592. [PMID: 33912935 DOI: 10.1242/dev.197111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 03/22/2021] [Indexed: 01/09/2023]
Abstract
In response to signals from the embryonic testis, the germ cell intrinsic factor NANOS2 coordinates a transcriptional program necessary for the differentiation of pluripotent-like primordial germ cells toward a unipotent spermatogonial stem cell fate. Emerging evidence indicates that genetic risk factors contribute to testicular germ cell tumor initiation by disrupting sex-specific differentiation. Here, using the 129.MOLF-Chr19 mouse model of testicular teratomas and a NANOS2 reporter allele, we report that the developmental phenotypes required for tumorigenesis, including failure to enter mitotic arrest, retention of pluripotency and delayed sex-specific differentiation, were exclusive to a subpopulation of germ cells failing to express NANOS2. Single-cell RNA sequencing revealed that embryonic day 15.5 NANOS2-deficient germ cells and embryonal carcinoma cells developed a transcriptional profile enriched for MYC signaling, NODAL signaling and primed pluripotency. Moreover, lineage-tracing experiments demonstrated that embryonal carcinoma cells arose exclusively from germ cells failing to express NANOS2. Our results indicate that NANOS2 is the nexus through which several genetic risk factors influence tumor susceptibility. We propose that, in the absence of sex specification, signals native to the developing testis drive germ cell transformation.
Collapse
Affiliation(s)
- Nicholas J Webster
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rebecca L Maywald
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Susan M Benton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Emily P Dawson
- Department of Cell Biology, New York University, New York, NY 10003, USA
| | - Oscar D Murillo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Emily L LaPlante
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Denise G Lanza
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
67
|
Role of stromal activin A in human pancreatic cancer and metastasis in mice. Sci Rep 2021; 11:7986. [PMID: 33846512 PMCID: PMC8042028 DOI: 10.1038/s41598-021-87213-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has extensive stromal involvement and remains one of the cancers with the highest mortality rates. Activin A has been implicated in colon cancer and its stroma but its role in the stroma of PDAC has not been elucidated. Activin A expression in cancer and stroma was assessed in human PDAC tissue microarrays (TMA). Activin A expression in human TMA is significantly higher in cancer samples, with expression in stroma correlated with shorter survival. Cultured pancreatic stellate cells (PSC) were found to secrete high levels of activin A resulting in PDAC cell migration that is abolished by anti-activin A neutralizing antibody. KPC mice treated with anti-activin A neutralizing antibody were evaluated for tumors, lesions and metastases quantified by immunohistochemistry. KPC mice with increased tumor burden express high plasma activin A. Treating KPC mice with an activin A neutralizing antibody does not reduce primary tumor size but decreases tumor metastases. From these data we conclude that PDAC patients with high activin A expression in stroma have a worse prognosis. PSCs secrete activin A, promoting increased PDAC migration. Inhibition of activin A in mice decreased metastases. Hence, stroma-rich PDAC patients might benefit from activin A inhibition.
Collapse
|
68
|
Zhang Y, Zhang Z, Chen P, Ma CY, Li C, Au TYK, Tam V, Peng Y, Wu R, Cheung KMC, Sham PC, Tse HF, Chan D, Leung VY, Cheah KSE, Lian Q. Directed Differentiation of Notochord-like and Nucleus Pulposus-like Cells Using Human Pluripotent Stem Cells. Cell Rep 2021; 30:2791-2806.e5. [PMID: 32101752 DOI: 10.1016/j.celrep.2020.01.100] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 10/15/2019] [Accepted: 01/28/2020] [Indexed: 12/26/2022] Open
Abstract
Intervertebral disc degeneration might be amenable to stem cell therapy, but the required cells are scarce. Here, we report the development of a protocol for directed in vitro differentiation of human pluripotent stem cells (hPSCs) into notochord-like and nucleus pulposus (NP)-like cells of the disc. The first step combines enhancement of ACTIVIN/NODAL and WNT and inhibition of BMP pathways. By day 5 of differentiation, hPSC-derived cells express notochordal cell characteristic genes. After activating the TGF-β pathway for an additional 15 days, qPCR, immunostaining, and transcriptome data show that a wide array of NP markers are expressed. Transcriptomically, the in vitro-derived cells become more like in vivo adolescent human NP cells, driven by a set of influential genes enriched with motifs bound by BRACHYURY and FOXA2, consistent with an NP cell-like identity. Transplantation of these NP-like cells attenuates fibrotic changes in a rat disc injury model of disc degeneration.
Collapse
Affiliation(s)
- Yuelin Zhang
- Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong; Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, Guangdong 510080, China
| | - Zhao Zhang
- Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong; Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, Guangdong 510080, China
| | - Peikai Chen
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Chui Yan Ma
- Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Cheng Li
- Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Tiffany Y K Au
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Vivian Tam
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Yan Peng
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Ron Wu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Kenneth Man Chee Cheung
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Pak C Sham
- Centre for PanorOmic Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Hung-Fat Tse
- Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Danny Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Victor Y Leung
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Kathryn S E Cheah
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong.
| | - Qizhou Lian
- Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong; Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, Guangdong 510080, China; The State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong.
| |
Collapse
|
69
|
Madsen RR, Longden J, Knox RG, Robin X, Völlmy F, Macleod KG, Moniz LS, Carragher NO, Linding R, Vanhaesebroeck B, Semple RK. NODAL/TGFβ signalling mediates the self-sustained stemness induced by PIK3CAH1047R homozygosity in pluripotent stem cells. Dis Model Mech 2021; 14:dmm048298. [PMID: 33514588 PMCID: PMC7969366 DOI: 10.1242/dmm.048298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/20/2021] [Indexed: 12/13/2022] Open
Abstract
Activating PIK3CA mutations are known 'drivers' of human cancer and developmental overgrowth syndromes. We recently demonstrated that the 'hotspot' PIK3CAH1047R variant exerts unexpected allele dose-dependent effects on stemness in human pluripotent stem cells (hPSCs). In this study, we combine high-depth transcriptomics, total proteomics and reverse-phase protein arrays to reveal potentially disease-related alterations in heterozygous cells, and to assess the contribution of activated TGFβ signalling to the stemness phenotype of homozygous PIK3CAH1047R cells. We demonstrate signalling rewiring as a function of oncogenic PI3K signalling strength, and provide experimental evidence that self-sustained stemness is causally related to enhanced autocrine NODAL/TGFβ signalling. A significant transcriptomic signature of TGFβ pathway activation in heterozygous PIK3CAH1047R was observed but was modest and was not associated with the stemness phenotype seen in homozygous mutants. Notably, the stemness gene expression in homozygous PIK3CAH1047R hPSCs was reversed by pharmacological inhibition of NODAL/TGFβ signalling, but not by pharmacological PI3Kα pathway inhibition. Altogether, this provides the first in-depth analysis of PI3K signalling in hPSCs and directly links strong PI3K activation to developmental NODAL/TGFβ signalling. This work illustrates the importance of allele dosage and expression when artificial systems are used to model human genetic disease caused by activating PIK3CA mutations. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Ralitsa R. Madsen
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
- Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
- The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| | - James Longden
- Biotech Research and Innovation Centre, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Theoretical Biophysics, Institute of Biology, Humboldt-Universität zu Berlin, 10115Berlin, Germany
| | - Rachel G. Knox
- Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
- The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| | - Xavier Robin
- Biotech Research and Innovation Centre, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Franziska Völlmy
- Biotech Research and Innovation Centre, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Kenneth G. Macleod
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Larissa S. Moniz
- University College London Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6BT, UK
| | - Neil O. Carragher
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Rune Linding
- Biotech Research and Innovation Centre, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Theoretical Biophysics, Institute of Biology, Humboldt-Universität zu Berlin, 10115Berlin, Germany
| | - Bart Vanhaesebroeck
- University College London Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6BT, UK
| | - Robert K. Semple
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| |
Collapse
|
70
|
Cermola F, D'Aniello C, Tatè R, De Cesare D, Martinez-Arias A, Minchiotti G, Patriarca EJ. Gastruloid Development Competence Discriminates Different States of Pluripotency. Stem Cell Reports 2021; 16:354-369. [PMID: 33482102 PMCID: PMC7878839 DOI: 10.1016/j.stemcr.2020.12.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 12/19/2020] [Accepted: 12/20/2020] [Indexed: 02/07/2023] Open
Abstract
Floating spheroidal aggregates of mouse embryonic stem cells can develop into polarized/elongated organoids, namely gastruloids. We set up a high-performing assay to measure gastruloid formation efficiency (GFE), and found that GFE decreases as pluripotency progresses from naive (GFE ≥ 95%) to primed (GFE = 0) state. Specifically, we show that primed EpiSCs fail to generate proper cell aggregates, while early-primed EpiLCs aggregate but eventually fail to develop into elongated gastruloids. Moreover, we characterized proline-induced cells (PiCs), a LIF-dependent reversible early-primed state of pluripotency, and show that PiCs are able to generate gastruloids (GFE ∼ 50%) and are also competent to differentiate into primordial germ cell-like cells. Thus, we propose the GFE assay as a valuable functional tool to discriminate different states of the pluripotency continuum.
Collapse
Affiliation(s)
- Federica Cermola
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics "A. Buzzati Traverso", CNR, Naples, Italy
| | - Cristina D'Aniello
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics "A. Buzzati Traverso", CNR, Naples, Italy
| | - Rosarita Tatè
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics "A. Buzzati Traverso", CNR, Naples, Italy
| | - Dario De Cesare
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics "A. Buzzati Traverso", CNR, Naples, Italy
| | | | - Gabriella Minchiotti
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics "A. Buzzati Traverso", CNR, Naples, Italy.
| | - Eduardo Jorge Patriarca
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics "A. Buzzati Traverso", CNR, Naples, Italy.
| |
Collapse
|
71
|
Xing Y, Liu J, Guo X, Liu H, Zeng W, Wang Y, Zhang C, Lu Y, He D, Ma S, He Y, Xing XH. Engineering organoid microfluidic system for biomedical and health engineering: A review. Chin J Chem Eng 2021. [DOI: 10.1016/j.cjche.2020.11.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
72
|
Dentate gyrus activin signaling mediates the antidepressant response. Transl Psychiatry 2021; 11:7. [PMID: 33414389 PMCID: PMC7791138 DOI: 10.1038/s41398-020-01156-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Antidepressants that target monoaminergic systems, such as selective serotonin reuptake inhibitors (SSRIs), are widely used to treat neuropsychiatric disorders including major depressive disorder, several anxiety disorders, and obsessive-compulsive disorder. However, these treatments are not ideal because only a subset of patients achieve remission. The reasons why some individuals remit to antidepressant treatments while others do not are unknown. Here, we developed a paradigm to assess antidepressant treatment resistance in mice. Exposure of male C57BL/6J mice to either chronic corticosterone administration or chronic social defeat stress induces maladaptive affective behaviors. Subsequent chronic treatment with the SSRI fluoxetine reverses these maladaptive affective behavioral changes in some, but not all, of the mice, permitting stratification into persistent responders and non-responders to fluoxetine. We found several differences in expression of Activin signaling-related genes between responders and non-responders in the dentate gyrus (DG), a region that is critical for the beneficial behavioral effects of fluoxetine. Enhancement of Activin signaling in the DG converted behavioral non-responders into responders to fluoxetine treatment more effectively than commonly used second-line antidepressant treatments, while inhibition of Activin signaling in the DG converted responders into non-responders. Taken together, these results demonstrate that the behavioral response to fluoxetine can be bidirectionally modified via targeted manipulations of the DG and suggest that molecular- and neural circuit-based modulations of DG may provide a new therapeutic avenue for more effective antidepressant treatments.
Collapse
|
73
|
Wang N, Weng J, Xia J, Zhu Y, Chen Q, Hu D, Zhang X, Sun R, Feng J, Minato N, Gong Y, Su L. SIPA1 enhances SMAD2/3 expression to maintain stem cell features in breast cancer cells. Stem Cell Res 2020; 49:102099. [PMID: 33296812 DOI: 10.1016/j.scr.2020.102099] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 09/30/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
SIPA1, a GTPase activating protein that negatively regulates Ras-related protein (Rap), is a potential modulator of tumor metastasis and recurrence. In this study, we first showed that SIPA1 facilitated the stemness features of breast cancer cells, such as of tumorsphere formation capability and the expression of stemness marker CD44. In addition, SIPA1 promoted the expression of four stemness-associated transcription factors through increasing the expression of SMAD2 and SMAD3 in vitro and in vivo. The stemness features were abolished by blocking the phosphorylation of SMAD3 with its specific inhibitor SIS3. Furthermore, SIPA1 decreased the breast cancer cell sensitivity to chemotherapy drugs. This effect was, however, competitively reversed by blocking the SMAD3 phosphorylation by SIS3 treatment in breast cancer cells. Taken together, SIPA1 promotes and sustains the stemness of breast cancer cells and their resistance to chemotherapy by increasing the expression of SMAD2 and SMAD3, and blocking SMAD3 phosphorylation could suppress the cancer cell stemness and increase the sensitivity to chemotherapy in breast cancer cells expressing a high level of SIPA1.
Collapse
Affiliation(s)
- Ning Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jun Weng
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jing Xia
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yangjin Zhu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qiongrong Chen
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Die Hu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xue Zhang
- Department of Breast Surgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Rui Sun
- Department of Oncology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430034, China
| | - Jueping Feng
- Department of Oncology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430034, China
| | - Nagahiro Minato
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yiping Gong
- Department of Breast Surgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China; Department of Breast Surgery, Hubei Cancer Hospital, Wuhan 430079, China.
| | - Li Su
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
74
|
Oppezzo A, Bourseguin J, Renaud E, Pawlikowska P, Rosselli F. Microphthalmia transcription factor expression contributes to bone marrow failure in Fanconi anemia. J Clin Invest 2020; 130:1377-1391. [PMID: 31877112 DOI: 10.1172/jci131540] [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: 07/03/2019] [Accepted: 12/11/2019] [Indexed: 12/20/2022] Open
Abstract
Hematopoietic stem cell (HSC) attrition is considered the key event underlying progressive BM failure (BMF) in Fanconi anemia (FA), the most frequent inherited BMF disorder in humans. However, despite major advances, how the cellular, biochemical, and molecular alterations reported in FA lead to HSC exhaustion remains poorly understood. Here, we demonstrated in human and mouse cells that loss-of-function of FANCA or FANCC, products of 2 genes affecting more than 80% of FA patients worldwide, is associated with constitutive expression of the transcription factor microphthalmia (MiTF) through the cooperative, unscheduled activation of several stress-signaling pathways, including the SMAD2/3, p38 MAPK, NF-κB, and AKT cascades. We validated the unrestrained Mitf expression downstream of p38 in Fanca-/- mice, which display hallmarks of hematopoietic stress, including loss of HSC quiescence, DNA damage accumulation in HSCs, and reduced HSC repopulation capacity. Importantly, we demonstrated that shRNA-mediated downregulation of Mitf expression or inhibition of p38 signaling rescued HSC quiescence and prevented DNA damage accumulation. Our data support the hypothesis that HSC attrition in FA is the consequence of defects in the DNA-damage response combined with chronic activation of otherwise transiently activated signaling pathways, which jointly prevent the recovery of HSC quiescence.
Collapse
Affiliation(s)
- Alessia Oppezzo
- CNRS UMR8200 Equipe Labellisée "La Ligue Contre le Cancer,".,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Orsay, France
| | - Julie Bourseguin
- CNRS UMR8200 Equipe Labellisée "La Ligue Contre le Cancer,".,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Orsay, France
| | - Emilie Renaud
- CNRS UMR8200 Equipe Labellisée "La Ligue Contre le Cancer,".,Gustave Roussy, Villejuif, France
| | - Patrycja Pawlikowska
- CNRS UMR8200 Equipe Labellisée "La Ligue Contre le Cancer,".,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Orsay, France
| | - Filippo Rosselli
- CNRS UMR8200 Equipe Labellisée "La Ligue Contre le Cancer,".,Gustave Roussy, Villejuif, France.,Université Paris-Saclay, Orsay, France
| |
Collapse
|
75
|
Matsumoto T, Chino H, Akiya M, Hashimura M, Yokoi A, Tochimoto M, Nakagawa M, Jiang Z, Saegusa M. Requirements of LEFTY and Nodal overexpression for tumor cell survival under hypoxia in glioblastoma. Mol Carcinog 2020; 59:1409-1419. [PMID: 33111989 DOI: 10.1002/mc.23265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 01/06/2023]
Abstract
Glioblastomas (GBM) contain numerous hypoxic foci associated with a rare fraction of glioma stem cells (GSCs). Left-right determination factor (LEFTY) and Nodal, members of the transforming growth factor β (TGF-β) superfamily, have glycogen synthase kinase 3β (GSK-3β) phosphorylation motifs and are linked with stemness in human malignancies. Herein, we investigated the roles of LEFTY and Nodal in GBM hypoxic foci. In clinical samples, significantly higher expression of LEFTY, Nodal, phospho (p) GSK-3β, pSmad2, and Nestin, as well as higher apoptotic and lower proliferation rates, were observed in nonpseudopalisading (non-Ps) perinecrotic lesions as compared to Ps and non-necrotic tumor lesions, with a positive correlation between LEFTY, Nodal, pGSK-3β, or pSmad2 scores. In KS-1, a GBM cell line that lacks endogenous Nodal expression, treatment with the hypoxic mimetic CoCl2 increased LEFTY, pGSK-3β, and pSmad2 levels, but decreased pAkt levels. Moreover, the promoter for LEFTY, but not Nodal, was activated by Smad2 or TGF-β1, suggesting that overexpression of LEFTY and Nodal may be due to Akt-independent GSK-3β inactivation, with or without cooperation of the TGF-β1/Smad2 axis. LEFTY and Nodal overexpression increased proliferation rates and reduced susceptibility to CoCl2 -induced apoptosis, and increased the expression of epithelial-mesenchymal transition (EMT)/GSC-related markers. An increased ALDH1high population and more efficient spheroid formation was also observed in LEFTY-overexpressing cells. These findings suggest that LEFTY and Nodal may contribute to cell survival in non-Ps GBM perinecrotic lesions, leading to alterations in apoptosis, proliferation, or EMT/GCS features.
Collapse
Affiliation(s)
- Toshihide Matsumoto
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Hiromi Chino
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Masashi Akiya
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Miki Hashimura
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Ako Yokoi
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Masataka Tochimoto
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Mayu Nakagawa
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Zesong Jiang
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Makoto Saegusa
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| |
Collapse
|
76
|
Influence of the TGF-β Superfamily on Osteoclasts/Osteoblasts Balance in Physiological and Pathological Bone Conditions. Int J Mol Sci 2020; 21:ijms21207597. [PMID: 33066607 PMCID: PMC7589189 DOI: 10.3390/ijms21207597] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 12/19/2022] Open
Abstract
The balance between bone forming cells (osteoblasts/osteocytes) and bone resorbing cells (osteoclasts) plays a crucial role in tissue homeostasis and bone repair. Several hormones, cytokines, and growth factors-in particular the members of the TGF-β superfamily such as the bone morphogenetic proteins-not only regulate the proliferation, differentiation, and functioning of these cells, but also coordinate the communication between them to ensure an appropriate response. Therefore, this review focuses on TGF-β superfamily and its influence on bone formation and repair, through the regulation of osteoclastogenesis, osteogenic differentiation of stem cells, and osteoblasts/osteoclasts balance. After introducing the main types of bone cells, their differentiation and cooperation during bone remodeling and fracture healing processes are discussed. Then, the TGF-β superfamily, its signaling via canonical and non-canonical pathways, as well as its regulation by Wnt/Notch or microRNAs are described and discussed. Its important role in bone homeostasis, repair, or disease is also highlighted. Finally, the clinical therapeutic uses of members of the TGF-β superfamily and their associated complications are debated.
Collapse
|
77
|
Hong M, Christ A, Christa A, Willnow TE, Krauss RS. Cdon mutation and fetal alcohol converge on Nodal signaling in a mouse model of holoprosencephaly. eLife 2020; 9:60351. [PMID: 32876567 PMCID: PMC7467722 DOI: 10.7554/elife.60351] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/10/2020] [Indexed: 02/07/2023] Open
Abstract
Holoprosencephaly (HPE), a defect in midline patterning of the forebrain and midface, arises ~1 in 250 conceptions. It is associated with predisposing mutations in the Nodal and Hedgehog (HH) pathways, with penetrance and expressivity graded by genetic and environmental modifiers, via poorly understood mechanisms. CDON is a multifunctional co-receptor, including for the HH pathway. In mice, Cdon mutation synergizes with fetal alcohol exposure, producing HPE phenotypes closely resembling those seen in humans. We report here that, unexpectedly, Nodal signaling is a major point of synergistic interaction between Cdon mutation and fetal alcohol. Window-of-sensitivity, genetic, and in vitro findings are consistent with a model whereby brief exposure of Cdon mutant embryos to ethanol during gastrulation transiently and partially inhibits Nodal pathway activity, with consequent effects on midline patterning. These results illuminate mechanisms of gene-environment interaction in a multifactorial model of a common birth defect.
Collapse
Affiliation(s)
- Mingi Hong
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Annabel Christ
- Max-Delbruck-Center for Molecular Medicine, Berlin, Germany
| | - Anna Christa
- Max-Delbruck-Center for Molecular Medicine, Berlin, Germany
| | | | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| |
Collapse
|
78
|
Optimizing the Use of iPSC-CMs for Cardiac Regeneration in Animal Models. Animals (Basel) 2020; 10:ani10091561. [PMID: 32887495 PMCID: PMC7552322 DOI: 10.3390/ani10091561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary In 2006, the first induced pluripotent stem cells were generated by reprogramming skin cells. Induced pluripotent stem cells undergo fast cell division, can differentiate into many different cell types, can be patient-specific, and do not raise ethical issues. Thus, they offer great promise as in vitro disease models, drug toxicity testing platforms, and for autologous tissue regeneration. Heart failure is one of the major causes of death worldwide. It occurs when the heart cannot meet the body’s metabolic demands. Induced pluripotent stem cells can be differentiated into cardiac myocytes, can form patches resembling native cardiac tissue, and can engraft to the damaged heart. However, despite correct host/graft coupling, most animal studies demonstrate an arrhythmogenicity of the engrafted tissue and variable survival. This is partially because of the heterogeneity and immaturity of the cells. New evidence suggests that by modulating induced pluripotent stem cells-cardiac myocytes (iPSC-CM) metabolism by switching substrates and changing metabolic pathways, you can decrease iPSC-CM heterogeneity and arrhythmogenicity. Novel culture methods and tissue engineering along with animal models of heart failure are needed to fully unlock the potential of cardiac myocytes derived from induced pluripotent stem cells for cardiac regeneration. Abstract Heart failure (HF) is a common disease in which the heart cannot meet the metabolic demands of the body. It mostly occurs in individuals 65 years or older. Cardiac transplantation is the best option for patients with advanced HF. High numbers of patient-specific cardiac myocytes (CMs) can be generated from induced pluripotent stem cells (iPSCs) and can possibly be used to treat HF. While some studies found iPSC-CMS can couple efficiently to the damaged heart and restore cardiac contractility, almost all found iPSC-CM transplantation is arrhythmogenic, thus hampering the use of iPSC-CMs for cardiac regeneration. Studies show that iPSC-CM cultures are highly heterogeneous containing atrial-, ventricular- and nodal-like CMs. Furthermore, they have an immature phenotype, resembling more fetal than adult CMs. There is an urgent need to overcome these issues. To this end, a novel and interesting avenue to increase CM maturation consists of modulating their metabolism. Combined with careful engineering and animal models of HF, iPSC-CMs can be assessed for their potential for cardiac regeneration and a cure for HF.
Collapse
|
79
|
Toyonaga T, Steinbach EC, Keith BP, Barrow JB, Schaner MR, Wolber EA, Beasley C, Huling J, Wang Y, Allbritton NL, Chaumont N, Sadiq TS, Koruda MJ, Jain A, Long MD, Barnes EL, Herfarth HH, Isaacs KL, Hansen JJ, Shanahan MT, Rahbar R, Furey TS, Sethupathy P, Sheikh SZ. Decreased Colonic Activin Receptor-Like Kinase 1 Disrupts Epithelial Barrier Integrity in Patients With Crohn's Disease. Cell Mol Gastroenterol Hepatol 2020; 10:779-796. [PMID: 32561494 PMCID: PMC7502566 DOI: 10.1016/j.jcmgh.2020.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Intestinal epithelial cell (IEC) barrier dysfunction is critical to the development of Crohn's disease (CD). However, the mechanism is understudied. We recently reported increased microRNA-31-5p (miR-31-5p) expression in colonic IECs of CD patients, but downstream targets and functional consequences are unknown. METHODS microRNA-31-5p target genes were identified by integrative analysis of RNA- and small RNA-sequencing data from colonic mucosa and confirmed by quantitative polymerase chain reaction in colonic IECs. Functional characterization of activin receptor-like kinase 1 (ACVRL1 or ALK1) in IECs was performed ex vivo using 2-dimensional cultured human primary colonic IECs. The impact of altered colonic ALK1 signaling in CD for the risk of surgery and endoscopic relapse was evaluated by a multivariate regression analysis and a Kaplan-Meier estimator. RESULTS ALK1 was identified as a target of miR-31-5p in colonic IECs of CD patients and confirmed using a 3'-untranslated region reporter assay. Activation of ALK1 restricted the proliferation of colonic IECs in a 5-ethynyl-2-deoxyuridine proliferation assay and down-regulated the expression of stemness-related genes. Activated ALK1 signaling increased colonic IEC differentiation toward colonocytes. Down-regulated ALK1 signaling was associated with increased stemness and decreased colonocyte-specific marker expression in colonic IECs of CD patients compared with healthy controls. Activation of ALK1 enhanced epithelial barrier integrity in a transepithelial electrical resistance permeability assay. Lower colonic ALK1 expression was identified as an independent risk factor for surgery and was associated with a higher risk of endoscopic relapse in CD patients. CONCLUSIONS Decreased colonic ALK1 disrupted colonic IEC barrier integrity and was associated with poor clinical outcomes in CD patients.
Collapse
Affiliation(s)
- Takahiko Toyonaga
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Erin C Steinbach
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Division of Rheumatology, Allergy and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Benjamin P Keith
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Genetics, Department of Biology, Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jasmine B Barrow
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Matthew R Schaner
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Elisabeth A Wolber
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Caroline Beasley
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jennifer Huling
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Yuli Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nancy L Allbritton
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nicole Chaumont
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Timothy S Sadiq
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Mark J Koruda
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Animesh Jain
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Millie D Long
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Edward L Barnes
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Hans H Herfarth
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kim L Isaacs
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jonathan J Hansen
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Michael T Shanahan
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Reza Rahbar
- Department of Surgery, Rex Healthcare of Wakefield, North Carolina
| | - Terrence S Furey
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Genetics, Department of Biology, Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Shehzad Z Sheikh
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
80
|
Al Hosni R, Shah M, Cheema U, Roberts HC, Luyten FP, Roberts SJ. Mapping human serum-induced gene networks as a basis for the creation of biomimetic periosteum for bone repair. Cytotherapy 2020; 22:424-435. [PMID: 32522398 DOI: 10.1016/j.jcyt.2020.03.434] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/21/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND The periosteum is a highly vascularized, collagen-rich tissue that plays a crucial role in directing bone repair. This is orchestrated primarily by its resident progenitor cell population. Indeed, preservation of periosteum integrity is critical for bone healing. Cells extracted from the periosteum retain their osteochondrogenic properties and as such are a promising basis for tissue engineering strategies for the repair of bone defects. However, the culture expansion conditions and the way in which the cells are reintroduced to the defect site are critical aspects of successful translation. Indeed, expansion in human serum and implantation on biomimetic materials has previously been shown to improve in vivo bone formation. AIM This study aimed to develop a protocol to allow for the expansion of human periosteum derived cells (hPDCs) in a biomimetic periosteal-like environment. METHODS The expansion conditions were defined through the investigation of the bioactive cues involved in augmenting hPDC proliferative and multipotency characteristics, based on transcriptomic analysis of cells cultured in human serum. RESULTS Master regulators of transcriptional networks were identified, and an optimized periosteum-derived growth factor cocktail (PD-GFC; containing β-estradiol, FGF2, TNFα, TGFβ, IGF-1 and PDGF-BB) was generated. Expansion of hPDCs in PD-GFC resulted in serum mimicry with regard to the cell morphology, proliferative capacity and chondrogenic differentiation. When incorporated into a three-dimensional collagen type 1 matrix and cultured in PD-GFC, the hPDCs migrated to the surface that represented the matrix topography of the periosteum cambium layer. Furthermore, gene expression analysis revealed a down-regulated WNT and TGFβ signature and an up-regulation of CREB, which may indicate the hPDCs are recreating their progenitor cell signature. CONCLUSION This study highlights the first stage in the development of a biomimetic periosteum, which may have applications in bone repair.
Collapse
Affiliation(s)
- Rawiya Al Hosni
- Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London, Stanmore, UK
| | - Mittal Shah
- Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London, Stanmore, UK
| | - Umber Cheema
- Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London, Stanmore, UK
| | - Helen C Roberts
- Department of Natural Sciences, Faculty of Science & Technology, Middlesex University, London, UK
| | - Frank P Luyten
- Skeletal Biology and Tissue Engineering Centre, Department of Development and Regeneration, KU Leuven, Leuven, Belgium and
| | - Scott J Roberts
- Department of Materials and Tissue, Institute of Orthopaedics and Musculoskeletal Science, University College London, Stanmore, UK; Skeletal Biology and Tissue Engineering Centre, Department of Development and Regeneration, KU Leuven, Leuven, Belgium and; Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK.
| |
Collapse
|
81
|
Panicker N, Coutman M, Lawlor-O’Neill C, Kahl RGS, Roselli S, Verrills NM. Ppp2r2a Knockout Mice Reveal That Protein Phosphatase 2A Regulatory Subunit, PP2A-B55α, Is an Essential Regulator of Neuronal and Epidermal Embryonic Development. Front Cell Dev Biol 2020; 8:358. [PMID: 32582689 PMCID: PMC7290052 DOI: 10.3389/fcell.2020.00358] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
The serine/threonine protein phosphatase 2A (PP2A) is a master regulator of the complex cellular signaling that occurs during all stages of mammalian development. PP2A is composed of a catalytic, a structural, and regulatory subunit, for which there are multiple isoforms. The association of specific regulatory subunits determines substrate specificity and localization of phosphatase activity, however, the precise role of each regulatory subunit in development is not known. Here we report the generation of the first knockout mouse for the Ppp2r2a gene, encoding the PP2A-B55α regulatory subunit, using CRISPR/Cas9. Heterozygous animals developed and grew as normal, however, homozygous knockout mice were not viable. Analysis of embryos at different developmental stages found a normal Mendelian ratio of Ppp2r2a-/- embryos at embryonic day (E) 10.5 (25%), but reduced Ppp2r2a-/- embryos at E14.5 (18%), and further reduced at E18.5 (10%). No live Ppp2r2a-/- pups were observed at birth. Ppp2r2a-/- embryos were significantly smaller than wild-type or heterozygous littermates and displayed a variety of neural defects such as exencephaly, spina bifida, and cranial vault collapse, as well as syndactyly and severe epidermal defects; all processes driven by growth and differentiation of the ectoderm. Ppp2r2a-/- embryos had incomplete epidermal barrier acquisition, associated with thin, poorly differentiated stratified epithelium with weak attachment to the underlying dermis. The basal keratinocytes in Ppp2r2a-/- embryos were highly disorganized, with reduced immunolabeling of integrins and basement membrane proteins, suggesting impaired focal adhesion and hemidesmosome assembly. The spinous and granular layers were thinner in the Ppp2r2a-/- embryos, with aberrant expression of adherens and tight junction associated proteins. The overlying stratum corneum was either absent or incomplete. Thus PP2A-B55α is an essential regulator of epidermal stratification, and is essential for ectodermal development during embryogenesis.
Collapse
Affiliation(s)
- Nikita Panicker
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation, University of Newcastle, Callaghan, NSW, Australia
- Hunter Cancer Research Alliance, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Melody Coutman
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation, University of Newcastle, Callaghan, NSW, Australia
- Hunter Cancer Research Alliance, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Charley Lawlor-O’Neill
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation, University of Newcastle, Callaghan, NSW, Australia
- Hunter Cancer Research Alliance, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Richard G. S. Kahl
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation, University of Newcastle, Callaghan, NSW, Australia
- Hunter Cancer Research Alliance, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Séverine Roselli
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation, University of Newcastle, Callaghan, NSW, Australia
- Hunter Cancer Research Alliance, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Nicole M. Verrills
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation, University of Newcastle, Callaghan, NSW, Australia
- Hunter Cancer Research Alliance, Hunter Medical Research Institute, New Lambton, NSW, Australia
| |
Collapse
|
82
|
Ghorbani-Dalini S, Azarpira N, Sangtarash MH, Soleimanpour-Lichaei HR, Yaghobi R, Lorzadeh S, Sabet A, Sarshar M, Al-Abdullah IH. Optimization of activin-A: a breakthrough in differentiation of human induced pluripotent stem cell into definitive endoderm. 3 Biotech 2020; 10:215. [PMID: 32355589 DOI: 10.1007/s13205-020-02215-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 04/18/2020] [Indexed: 01/09/2023] Open
Abstract
The first step in differentiation of pluripotent stem cell toward endoderm-derived cell/organ is differentiation to definitive endoderm (DE) which is the central issue in developmental biology. Based on several evidences, we hypothesized that activin-A optimization as well as replacement of fetal bovine serum (FBS) with knockout serum replacement (KSR) is important for differentiation of induced pluripotent stem cell (iPSC) line into DE. Therefore, a stepwise differentiation protocol was applied on R1-hiPSC1 cell line. At first, activin-A concentration (30, 50, 70 and 100 ng/ml) was optimized. Then, substitution of FBS with KSR was evaluated across four treatment groups. The amount of differentiation of iPSC toward DE was determined by quantitative gene expression analyses of pluripotency (NANOG and OCT4), definitive endoderm (SOX17 and FOXA2) and endoderm-derived organs (PDX1, NEUROG3, and PAX6). Based on gene expression analyses, the more decrease in concentrations of activin-A can increase the differentiation of iPSC into DE, therefore, 30 ng/ml activin-A was chosen as the best concentration for the differentiation of R1-hiPSC1 line toward endoderm-derived organ. Moreover, complete replacement of FBS with gradually increased KSR improved the differentiation of iPSC toward DE. For this reason, the addition of 0% KSR at day 1, 0.2% at day 2 and 2% for the next 3 days was the best optimal protocol of the differentiation of iPSC toward DE. Overall, our results demonstrate that optimization of activin-A is important for differentiation of iPSC line. Furthermore, the replacement of FBS with KSR can improve the efficiency of iPSC differentiation toward DE.
Collapse
Affiliation(s)
| | - Negar Azarpira
- 1Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Hamid Reza Soleimanpour-Lichaei
- 3Department of Stem Cells and Regenerative Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Ramin Yaghobi
- 1Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahrokh Lorzadeh
- 1Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alice Sabet
- 1Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Meysam Sarshar
- 4Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur Italia-Cenci Bolognetti Foundation, 00185 Rome, Italy
- 5Microbiology Research Center (MRC), Pasteur Institute of Iran, 1316943551 Tehran, Iran
| | - Ismail H Al-Abdullah
- 6Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, USA
| |
Collapse
|
83
|
Williams ML, Solnica-Krezel L. Nodal and planar cell polarity signaling cooperate to regulate zebrafish convergence and extension gastrulation movements. eLife 2020; 9:54445. [PMID: 32319426 PMCID: PMC7250581 DOI: 10.7554/elife.54445] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
During vertebrate gastrulation, convergence and extension (C and E) of the primary anteroposterior (AP) embryonic axis is driven by polarized mediolateral (ML) cell intercalations and is influenced by AP axial patterning. Nodal signaling is essential for patterning of the AP axis while planar cell polarity (PCP) signaling polarizes cells with respect to this axis, but how these two signaling systems interact during C and E is unclear. We find that the neuroectoderm of Nodal-deficient zebrafish gastrulae exhibits reduced C and E cell behaviors, which require Nodal signaling in both cell- and non-autonomous fashions. PCP signaling is partially active in Nodal-deficient embryos and its inhibition exacerbates their C and E defects. Within otherwise naïve zebrafish blastoderm explants, however, Nodal induces C and E in a largely PCP-dependent manner, arguing that Nodal acts both upstream of and in parallel with PCP during gastrulation to regulate embryonic axis extension cooperatively.
Collapse
Affiliation(s)
- Margot Lk Williams
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, United States
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, United States
| |
Collapse
|
84
|
Xiong C, Wang M, Ling W, Xie D, Chu X, Li Y, Huang Y, Li T, Otieno E, Qiu X, Xiao X. Advances in Isolation and Culture of Chicken Embryonic Stem Cells In Vitro. Cell Reprogram 2020; 22:43-54. [PMID: 32150690 DOI: 10.1089/cell.2019.0080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chicken embryonic stem cells (cESCs) isolated from the egg at the stage X hold great promise for cell therapy, tissue engineering, pharmaceutical, and biotechnological applications. They are considered to be pluripotent cells with the capacity to self-renewal and differentiate into specialized cells. However, long-term maintenance of cESCs cannot be realized now, which impedes the establishment of cESC line and limits their applications. Therefore, the separation locations, isolation methods, and culture conditions especially the supplements and action mechanisms of cytokines, including leukemia inhibitory factor, fibroblast growth factor, transforming growth factor beta, bone morphogenic protein, and activin for cESCs in vitro, have been reviewed here. These defined strategies will contribute to identify the key mechanism on the self-renewal of cESCs, facilitate to optimize system that supports the derivation and longtime maintenance of cESCs, establish the cESC line, and develop the biobank of genetic resources in chicken.
Collapse
Affiliation(s)
- Chunxia Xiong
- Department of Veterinary Medicine, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Mingyu Wang
- Department of Veterinary Medicine, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Wenhui Ling
- Department of Veterinary Medicine, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Dengfeng Xie
- Department of Veterinary Medicine, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Xinyue Chu
- Department of Veterinary Medicine, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yunxin Li
- Department of Veterinary Medicine, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yun Huang
- Department of Veterinary Medicine, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Tong Li
- Department of Veterinary Medicine, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Edward Otieno
- Department of Veterinary Medicine, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Xiaoyan Qiu
- Department of Veterinary Medicine, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Xiong Xiao
- Department of Veterinary Medicine, College of Animal Science and Technology, Southwest University, Chongqing, China
| |
Collapse
|
85
|
Xu X, Zhou X, Gao C, Cao L, Zhang Y, Hu X, Cui Y. Nodal promotes the malignancy of non-small cell lung cancer (NSCLC) cells via activation of NF-κB/IL-6 signals. Biol Chem 2020; 400:777-785. [PMID: 30699065 DOI: 10.1515/hsz-2018-0392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/16/2019] [Indexed: 01/09/2023]
Abstract
Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer deaths worldwide. Understanding the mechanisms responsible for the malignancy of NSCLC cells is important for therapy and drug development. Nodal, an important embryonic morphogen, has been reported to modulate tumorigenesis. We found that Nodal can trigger the proliferation of NSCLC cells and decrease the sensitivity to doxorubicin (Dox) and cisplatin (CDDP) treatment. Targeted inhibition of Nodal can suppress the proliferation of NSCLC cells. Among the measured cytokines, Nodal can increase the expression of interleukin-6 (IL-6) and vascular endothelial growth factor A (VEGFA) in NSCLC cells. Inhibition of IL-6, while not VEGFA, attenuated Nodal induced cell proliferation, suggesting the essential roles of IL-6 in Nodal induced malignancy of NSCLC cells. Nodal can trigger the phosphorylation, nuclear translocation and transcriptional activities of p65, the key signal transducer of NF-κB. This was due to the fact that Nodal can increase the phosphorylation of IKKβ/IκBα. The inhibitor of IKKβ abolished Nodal induced activation of p65 and expression of IL-6. Collectively, we found that Nodal can increase the proliferation and decrease chemosensitivity of NSCLC cells via regulation of NF-κB/IL-6 signals. It indicated that Nodal might be a potential therapeutic target for NSCLC treatment.
Collapse
Affiliation(s)
- Xiaohui Xu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China
| | - Xiaoyun Zhou
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China
| | - Chao Gao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China
| | - Lei Cao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China
| | - Ye Zhang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China
| | - Xue Hu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China
| | - Yushang Cui
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Shuaifuyuan No. 1, Dongcheng District, Beijing 100730, China
| |
Collapse
|
86
|
|
87
|
Kaminska B, Cyranowski S. Recent Advances in Understanding Mechanisms of TGF Beta Signaling and Its Role in Glioma Pathogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1202:179-201. [PMID: 32034714 DOI: 10.1007/978-3-030-30651-9_9] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Transforming growth factor beta (TGF-β) signaling is involved in the regulation of proliferation, differentiation and survival/or apoptosis of many cells, including glioma cells. TGF-β acts via specific receptors activating multiple intracellular pathways resulting in phosphorylation of receptor-regulated Smad2/3 proteins that associate with the common mediator, Smad4. Such complex translocates to the nucleus, binds to DNA and regulates transcription of many genes. Furthermore, TGF-β-activated kinase-1 (TAK1) is a component of TGF-β signaling and activates mitogen-activated protein kinase (MAPK) cascades. Negative regulation of TGF-β/Smad signaling may occur through the inhibitory Smad6/7. While genetic alterations in genes related to TGF-β signaling are relatively rare in gliomas, the altered expression of those genes is a frequent event. The increased expression of TGF-β1-3 correlates with a degree of malignancy of human gliomas. TGF-β may contribute to tumor pathogenesis in many ways: by direct support of tumor growth, by maintaining self-renewal of glioma initiating stem cells and inhibiting anti-tumor immunity. Glioma initiating cells are dedifferentiated cells that retain many stem cell-like properties, play a role in tumor initiation and contribute to its recurrence. TGF-β1,2 stimulate expression of the vascular endothelial growth factor as well as the plasminogen activator inhibitor and some metalloproteinases that are involved in vascular remodeling, angiogenesis and degradation of the extracellular matrix. Inhibitors of TGF-β signaling reduce viability and invasion of gliomas in animal models and show a great promise as novel, potential anti-tumor therapeutics.
Collapse
Affiliation(s)
- Bozena Kaminska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland. .,Postgraduate School of Molecular Medicine, Warsaw Medical University, Warsaw, Poland.
| | - Salwador Cyranowski
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.,Postgraduate School of Molecular Medicine, Warsaw Medical University, Warsaw, Poland
| |
Collapse
|
88
|
GREB1 induced by Wnt signaling promotes development of hepatoblastoma by suppressing TGFβ signaling. Nat Commun 2019; 10:3882. [PMID: 31462641 PMCID: PMC6713762 DOI: 10.1038/s41467-019-11533-x] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 07/18/2019] [Indexed: 12/16/2022] Open
Abstract
The β-catenin mutation is frequently observed in hepatoblastoma (HB), but the underlying mechanism by which Wnt/β-catenin signaling induces HB tumor formation is unknown. Here we show that expression of growth regulation by estrogen in breast cancer 1 (GREB1) depends on Wnt/β-catenin signaling in HB patients. GREB1 is localized to the nucleus where it binds Smad2/3 in a competitive manner with p300 and inhibits TGFβ signaling, thereby promoting HepG2 HB cell proliferation. Forced expression of β-catenin, YAP, and c-Met induces HB-like mouse liver tumor (BYM mice), with an increase in GREB1 expression and HB markers. Depletion of GREB1 strongly suppresses marker gene expression and HB-like liver tumorigenesis, and instead enhances TGFβ signaling in BYM mice. Furthermore, antisense oligonucleotides for GREB1 suppress the formation of HepG2 cell-induced tumors and HB-like tumors in vivo. We propose that GREB1 is a target molecule of Wnt/β-catenin signaling and required for HB progression. The mechanisms promoting hepatoblastoma (HB) progression through Wnt/β-catenin signaling are unclear. Here, the authors show that the Wnt/ β-catenin axis induces GREB1 expression and nuclear localization, and suppresses TGFβ pathway, and propose GREB1 as a therapeutic target in HB.
Collapse
|
89
|
Human Pluripotent Stem Cell-Derived Endoderm for Modeling Development and Clinical Applications. Cell Stem Cell 2019; 22:485-499. [PMID: 29625066 DOI: 10.1016/j.stem.2018.03.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The liver, lung, pancreas, and digestive tract all originate from the endoderm germ layer, and these vital organs are subject to many life-threatening diseases affecting millions of patients. However, primary cells from endodermal organs are often difficult to grow in vitro. Human pluripotent stem cells thus hold great promise for generating endoderm cells and their derivatives as tools for the development of new therapeutics against a variety of global healthcare challenges. Here we describe recent advances in methods for generating endodermal cell types from human pluripotent stem cells and their use for disease modeling and cell-based therapy.
Collapse
|
90
|
Roudebush C, Catala-Valentin A, Andl T, Le Bras GF, Andl CD. Activin A-mediated epithelial de-differentiation contributes to injury repair in an in vitro gastrointestinal reflux model. Cytokine 2019; 123:154782. [PMID: 31369967 DOI: 10.1016/j.cyto.2019.154782] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 02/08/2023]
Abstract
Reflux esophagitis is a result of esophageal exposure to acid and bile during episodes of gastroesophageal reflux. Aside from chemical injury to the esophageal epithelium, it has been shown that acid and bile induce cytokine-mediated injury by stimulating the release of pro-inflammatory cytokines. During the repair and healing process following reflux injury, the squamous esophageal cells are replaced with a columnar epithelium causing Barrett's metaplasia, which predisposes patients to esophageal adenocarcinoma. We identified a novel player in gastroesophageal reflux injury, the TGFβ family member Activin A (ActA), which is a known regulator of inflammation and tissue repair. In this study, we show that in response to bile salt and acidified media (pH 4) exposure, emulating the milieu to which the distal esophagus is exposed during gastroesophageal reflux, long-term treated, tolerant esophageal keratinocytes exhibit increased ActA secretion and a pro-inflammatory cytokine signature. Furthermore, we noted increased motility and expression of the stem cell markers SOX9, LGR5 and DCLK1 supporting the notion that repair mechanisms were activated in the bile salt/acid-tolerant keratinocytes. Additionally, these experiments demonstrated that de-differentiation as characterized by the induction of YAP1, FOXO3 and KRT17 was altered by ActA/TGFβ signaling. Collectively, our results suggest a pivotal role for ActA in the inflammatory GERD environment by modulating esophageal tissue repair and de-differentiation.
Collapse
Affiliation(s)
- Cedric Roudebush
- Burnett School of Biomedical Sciences, University of Central Florida, 4110 Libra Dr., BMS, Building 20, rm 223, Orlando, FL 32816, United States
| | - Alma Catala-Valentin
- Burnett School of Biomedical Sciences, University of Central Florida, 4110 Libra Dr., BMS, Building 20, rm 223, Orlando, FL 32816, United States
| | - Thomas Andl
- Burnett School of Biomedical Sciences, University of Central Florida, 4110 Libra Dr., BMS, Building 20, rm 223, Orlando, FL 32816, United States
| | - Gregoire F Le Bras
- Burnett School of Biomedical Sciences, University of Central Florida, 4110 Libra Dr., BMS, Building 20, rm 223, Orlando, FL 32816, United States
| | - Claudia D Andl
- Burnett School of Biomedical Sciences, University of Central Florida, 4110 Libra Dr., BMS, Building 20, rm 223, Orlando, FL 32816, United States.
| |
Collapse
|
91
|
The Functions of Long Non-Coding RNA during Embryonic Cardiovascular Development and Its Potential for Diagnosis and Treatment of Congenital Heart Disease. J Cardiovasc Dev Dis 2019; 6:jcdd6020021. [PMID: 31159401 PMCID: PMC6616656 DOI: 10.3390/jcdd6020021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 12/17/2022] Open
Abstract
Congenital heart disease (CHD) arises due to errors during the embryonic development of the heart, a highly regulated process involving an interplay between cell-intrinsic transcription factor expression and intercellular signalling mediated by morphogens. Emerging evidence indicates that expression of these protein-coding genes is controlled by a plethora of previously unappreciated non-coding RNAs operating in complex feedback-control circuits. In this review, we consider the contribution of long non-coding RNA (lncRNA) to embryonic cardiovascular development before discussing applications to CHD diagnostics and therapeutics. We discuss the process of lineage restriction during cardiovascular progenitor cell differentiation, as well as the subsequent patterning of the cardiogenic progenitor fields, taking as an example the regulation of NODAL signalling in left-right patterning of the heart. lncRNA are a highly versatile group. Nuclear lncRNA can target specific genomic sequences and recruit chromatin remodelling complexes. Some nuclear lncRNA are transcribed from enhancers and regulate chromatin looping. Cytoplasmic lncRNA act as endogenous competitors for micro RNA, as well as binding and sequestering signalling proteins. We discuss features of lncRNA that limit their study by conventional methodology and suggest solutions to these problems.
Collapse
|
92
|
Polonio-Alcalá E, Rabionet M, Ruiz-Martínez S, Ciurana J, Puig T. Three-Dimensional Manufactured Supports for Breast Cancer Stem Cell Population Characterization. Curr Drug Targets 2019; 20:839-851. [DOI: 10.2174/1389450120666181122113300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/03/2018] [Accepted: 11/07/2018] [Indexed: 12/23/2022]
Abstract
Breast Cancer (BC) is the most common cancer among women and the second cause of female death for cancer. When the tumor is not correctly eradicated, there is a high relapse risk and incidence of metastasis. Breast Cancer Stem Cells (BCSCs) are responsible for initiating tumors and are resistant to current anticancer therapies being in part responsible for tumor relapse and metastasis. The study of BCSCs is limited due to their low percentage within both tumors and established cell models. Hence, three-dimensional (3D) supports are presented as an interesting tool to keep the stem-like features in 3D cell culture. In this review, several 3D culture systems are discussed. Moreover, scaffolds are presented as a tool to enrich in BCSCs in order to find new specific therapeutic strategies against this malignant subpopulation. Anticancer treatments focused on BCSCs could be useful for BC patients, with particular interest in those that progress to current therapies.
Collapse
Affiliation(s)
- Emma Polonio-Alcalá
- New Therapeutic Targets Laboratory (TargetsLab), Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Marc Rabionet
- New Therapeutic Targets Laboratory (TargetsLab), Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Santiago Ruiz-Martínez
- New Therapeutic Targets Laboratory (TargetsLab), Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Joaquim Ciurana
- Product, Process and Production Engineering Research Group (GREP), Department of Mechanical Engineering and Industrial Construction, University of Girona, Girona, Spain
| | - Teresa Puig
- New Therapeutic Targets Laboratory (TargetsLab), Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| |
Collapse
|
93
|
Wu J, Cheng P, Huang Z, Tan Q, Qu Y. Nodal increases the malignancy of childhood neuroblastoma cells via regulation of Zeb1. Biofactors 2019; 45:355-363. [PMID: 30985990 DOI: 10.1002/biof.1505] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 02/28/2019] [Indexed: 12/14/2022]
Abstract
Neuroblastoma (NB) is one of the most common malignant tumors derived from pluripotent cells of the neural crest. Nodal is an important embryonic morphogen which can re-express in cancer cells. The roles of Nodal in the progression of NB are not illustrated. Our present study reveals that Nodal is upregulated in NB cells and tissues. Targeted inhibition of Nodal can suppress the in vitro migration and invasion of NB cells while increase its chemo-sensitivity to doxorubicin (Dox) treatment. Nodal positively regulates the expression of Zeb1, one well-known transcription factors of epithelial to mesenchymal transition (EMT) of cancer cells. Knockdown of Zeb1 can attenuate Nodal-induced malignancy of NB cells. Mechanistically, Nodal increases the protein stability of Zeb1 while has no effect on its mRNA expression. It is due to that Nodal can increase the expression of Ataxia telangiectasia mutated kinase (ATM), which can phosphorylate and stabilize Zeb1 in cancer cells. Collectively, our data revealed that Nodal can increase the malignancy of NB cells via increasing the expression of Zeb1. It suggests that targeted inhibition of Nodal might be a potential therapy approach for NB treatment. © 2019 BioFactors, 45(3):355-363, 2019.
Collapse
Affiliation(s)
- Jingfang Wu
- Department of Pediatrics, The Affiliated Hospital of Jining Medical University, Jining City, Shandong Province, China
| | - Panpan Cheng
- Lab of Hematology Department, The Affiliated Hospital of Jining Medical University, Jining City, Shandong Province, China
| | - Zongxuan Huang
- Department of Pediatrics, The Affiliated Hospital of Jining Medical University, Jining City, Shandong Province, China
| | - Qingshi Tan
- Department of Emergency, The Affiliated Hospital of Jining Medical University, Jining City, Shandong Province, China
| | - Yuhua Qu
- Department of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou City, Guangdong Province, China
| |
Collapse
|
94
|
Madsen RR, Knox RG, Pearce W, Lopez S, Mahler-Araujo B, McGranahan N, Vanhaesebroeck B, Semple RK. Oncogenic PIK3CA promotes cellular stemness in an allele dose-dependent manner. Proc Natl Acad Sci U S A 2019; 116:8380-8389. [PMID: 30948643 PMCID: PMC6486754 DOI: 10.1073/pnas.1821093116] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The PIK3CA gene, which encodes the p110α catalytic subunit of PI3 kinase (PI3K), is mutationally activated in cancer and in overgrowth disorders known as PIK3CA-related overgrowth spectrum (PROS). To determine the consequences of genetic PIK3CA activation in a developmental context of relevance to both PROS and cancer, we engineered isogenic human induced pluripotent stem cells (iPSCs) with heterozygous or homozygous knockin of PIK3CAH1047R While heterozygous iPSCs remained largely similar to wild-type cells, homozygosity for PIK3CAH1047R caused widespread, cancer-like transcriptional remodeling, partial loss of epithelial morphology, up-regulation of stemness markers, and impaired differentiation to all three germ layers in vitro and in vivo. Genetic analysis of PIK3CA-associated cancers revealed that 64% had multiple oncogenic PIK3CA copies (39%) or additional PI3K signaling pathway-activating "hits" (25%). This contrasts with the prevailing view that PIK3CA mutations occur heterozygously in cancer. Our findings suggest that a PI3K activity threshold determines pathological consequences of oncogenic PIK3CA activation and provide insight into the specific role of this pathway in human pluripotent stem cells.
Collapse
Affiliation(s)
- Ralitsa R Madsen
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
- National Institute for Health Research, Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, United Kingdom
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Rachel G Knox
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
- National Institute for Health Research, Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, United Kingdom
| | - Wayne Pearce
- University College London Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Saioa Lopez
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London WC1E 6DD, United Kingdom
- Cancer Genome Evolution Research Group, University College London Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Betania Mahler-Araujo
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
- Histopathology Department, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London WC1E 6DD, United Kingdom
- Cancer Genome Evolution Research Group, University College London Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Bart Vanhaesebroeck
- University College London Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Robert K Semple
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, United Kingdom;
- National Institute for Health Research, Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, United Kingdom
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| |
Collapse
|
95
|
Pacitti D, Privolizzi R, Bax BE. Organs to Cells and Cells to Organoids: The Evolution of in vitro Central Nervous System Modelling. Front Cell Neurosci 2019; 13:129. [PMID: 31024259 PMCID: PMC6465581 DOI: 10.3389/fncel.2019.00129] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/14/2019] [Indexed: 02/05/2023] Open
Abstract
With 100 billion neurons and 100 trillion synapses, the human brain is not just the most complex organ in the human body, but has also been described as "the most complex thing in the universe." The limited availability of human living brain tissue for the study of neurogenesis, neural processes and neurological disorders has resulted in more than a century-long strive from researchers worldwide to model the central nervous system (CNS) and dissect both its striking physiology and enigmatic pathophysiology. The invaluable knowledge gained with the use of animal models and post mortem human tissue remains limited to cross-species similarities and structural features, respectively. The advent of human induced pluripotent stem cell (hiPSC) and 3-D organoid technologies has revolutionised the approach to the study of human brain and CNS in vitro, presenting great potential for disease modelling and translational adoption in drug screening and regenerative medicine, also contributing beneficially to clinical research. We have surveyed more than 100 years of research in CNS modelling and provide in this review an historical excursus of its evolution, from early neural tissue explants and organotypic cultures, to 2-D patient-derived cell monolayers, to the latest development of 3-D cerebral organoids. We have generated a comprehensive summary of CNS modelling techniques and approaches, protocol refinements throughout the course of decades and developments in the study of specific neuropathologies. Current limitations and caveats such as clonal variation, developmental stage, validation of pluripotency and chromosomal stability, functional assessment, reproducibility, accuracy and scalability of these models are also discussed.
Collapse
Affiliation(s)
- Dario Pacitti
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London, United Kingdom
- College of Medicine and Health, St Luke’s Campus, University of Exeter, Exeter, United Kingdom
| | - Riccardo Privolizzi
- Gene Transfer Technology Group, Institute for Women’s Health, University College London, London, United Kingdom
| | - Bridget E. Bax
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London, United Kingdom
| |
Collapse
|
96
|
Zhou Q, Xia S, Guo F, Hu F, Wang Z, Ni Y, Wei T, Xiang H, Shang D. Transforming growth factor-β in pancreatic diseases: Mechanisms and therapeutic potential. Pharmacol Res 2019; 142:58-69. [PMID: 30682425 DOI: 10.1016/j.phrs.2019.01.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/27/2018] [Accepted: 01/18/2019] [Indexed: 12/16/2022]
Abstract
Pancreatic diseases, such as acute pancreatitis, chronic pancreatitis, and pancreatic cancer, are common gastrointestinal diseases resulting in the development of local and systemic complications with a high risk of death. Numerous studies have examined pancreatic diseases over the past few decades; however, the pathogenesis remains unclear, and there is a lack of effective treatment options. Recently, emerging evidence has suggested that transforming growth factor beta (TGF-β) exerts controversial functions in apoptosis, inflammatory responses, and carcinogenesis, indicating its complex role in the pathogenesis of pancreas-associated disease. Therefore, a further understanding of relevant TGF-β signalling will provide new ideas and potential therapeutic targets for preventing disease progression. This is the first systematic review of recent data from animal and human clinical studies focusing on TGF-β signalling in pancreas damage and diseases. This information may aid in the development of therapeutic agents for regulating TGF-β in this pathology to prevent or treat pancreatic diseases.
Collapse
Affiliation(s)
- Qi Zhou
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China; Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shilin Xia
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fangyue Guo
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Fenglin Hu
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Zhizhou Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yujia Ni
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Tianfu Wei
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Hong Xiang
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Dong Shang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China; Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
97
|
Wei X, Guo J, Li Q, Jia Q, Jing Q, Li Y, Zhou B, Chen J, Gao S, Zhang X, Jia M, Niu C, Yang W, Zhi X, Wang X, Yu D, Bai L, Wang L, Na J, Zou Y, Zhang J, Zhang S, Meng D. Bach1 regulates self-renewal and impedes mesendodermal differentiation of human embryonic stem cells. SCIENCE ADVANCES 2019; 5:eaau7887. [PMID: 30891497 PMCID: PMC6415956 DOI: 10.1126/sciadv.aau7887] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 01/30/2019] [Indexed: 05/03/2023]
Abstract
The transcription factor BTB and CNC homology 1 (Bach1) is expressed in the embryos of mice, but whether Bach1 regulates the self-renewal and early differentiation of human embryonic stem cells (hESCs) is unknown. We report that the deubiquitinase ubiquitin-specific processing protease 7 (Usp7) is a direct target of Bach1, that Bach1 interacts with Nanog, Sox2, and Oct4, and that Bach1 facilitates their deubiquitination and stabilization via the recruitment of Usp7, thereby maintaining stem cell identity and self-renewal. Bach1 also interacts with polycomb repressive complex 2 (PRC2) and represses mesendodermal gene expression by recruiting PRC2 to the genes' promoters. The loss of Bach1 in hESCs promotes differentiation toward the mesendodermal germ layers by reducing the occupancy of EZH2 and H3K27me3 in mesendodermal gene promoters and by activating the Wnt/β-catenin and Nodal/Smad2/3 signaling pathways. Our study shows that Bach1 is a key determinant of pluripotency, self-renewal, and lineage specification in hESCs.
Collapse
Affiliation(s)
- Xiangxiang Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jieyu Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Qinhan Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Qianqian Jia
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Qing Jing
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Li
- The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiayu Chen
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Shaorong Gao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xinyue Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Mengping Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Cong Niu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Wenlong Yang
- Department of Cardiology, Zhongshan Hospital, Shanghai Cardiovascular Medical Center, Shanghai Institute of Cardiovascular Diseases, Institute of Pan-vascular Medicine, Fudan University, Shanghai 200032, China
| | - Xiuling Zhi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xinhong Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Dian Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Lufeng Bai
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Lin Wang
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Jie Na
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yunzeng Zou
- Department of Cardiology, Zhongshan Hospital, Shanghai Cardiovascular Medical Center, Shanghai Institute of Cardiovascular Diseases, Institute of Pan-vascular Medicine, Fudan University, Shanghai 200032, China
| | - Jianyi Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Shuning Zhang
- Department of Cardiology, Zhongshan Hospital, Shanghai Cardiovascular Medical Center, Shanghai Institute of Cardiovascular Diseases, Institute of Pan-vascular Medicine, Fudan University, Shanghai 200032, China
- Corresponding author. (D.M.); (S.Z.)
| | - Dan Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Corresponding author. (D.M.); (S.Z.)
| |
Collapse
|
98
|
Wang X, Tang P, Guo F, Zhang M, Yan Y, Huang M, Chen Y, Zhang L, Zhang L. mDia1 and Cdc42 Regulate Activin B-Induced Migration of Bone Marrow-Derived Mesenchymal Stromal Cells. Stem Cells 2019; 37:150-162. [PMID: 30358011 PMCID: PMC7379979 DOI: 10.1002/stem.2924] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 08/23/2018] [Accepted: 08/28/2018] [Indexed: 12/20/2022]
Abstract
In a previous study, we have shown that Activin B is a potent chemoattractant for bone marrow-derived mesenchymal stromal cells (BMSCs). As such, the combination of Activin B and BMSCs significantly accelerated rat skin wound healing. In another study, we showed that RhoA activation plays a key role in Activin B-induced BMSC migration. However, the role of the immediate downstream effectors of RhoA in this process is unclear. Here, we demonstrated that mammalian homolog of Drosophila diaphanous-1 (mDia1), a downstream effector of RhoA, exerts a crucial function in Activin B-induced BMSC migration by promoting membrane ruffling, microtubule morphology, and adhesion signaling dynamics. Furthermore, we showed that Activin B does not change Rac1 activity but increases Cdc42 activity in BMSCs. Inactivation of Cdc42 inhibited Activin B-stimulated Golgi reorientation and the cell migration of BMSCs. Furthermore, knockdown of mDia1 affected Activin B-induced BMSC-mediated wound healing in vivo. In conclusion, this study demonstrated that the RhoA-mDia1 and Cdc42 pathways regulate Activin B-induced BMSC migration. This study may help to optimize clinical MSC-based transplantation strategies to promote skin wound healing. Stem Cells 2019;37:150-162.
Collapse
Affiliation(s)
- Xueer Wang
- Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Pei Tang
- Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer BiologyChildren's Hospital Research FoundationCincinnatiOhioUSA
| | - Min Zhang
- Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Yuan Yan
- Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Mianbo Huang
- Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Yinghua Chen
- Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Lu Zhang
- Guangdong Provincial Key Laboratory of Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Lin Zhang
- Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| |
Collapse
|
99
|
Sweeney M, Foldes G. It Takes Two: Endothelial-Perivascular Cell Cross-Talk in Vascular Development and Disease. Front Cardiovasc Med 2018; 5:154. [PMID: 30425990 PMCID: PMC6218412 DOI: 10.3389/fcvm.2018.00154] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/10/2018] [Indexed: 12/26/2022] Open
Abstract
The formation of new blood vessels is a crucial step in the development of any new tissue both during embryogenesis and in vitro models as without sufficient perfusion the tissue will be unable to grow beyond the size where nutrition and oxygenation can be managed by diffusion alone. Endothelial cells are the primary building block of blood vessels and are capable of forming tube like structures independently however they are unable to independently form functional vasculature which is capable of conducting blood flow. This requires support from other structures including supporting perivascular cells and the extracellular matrix. The crosstalk between endothelial cells and perivascular cells is vital in regulating vasculogenesis and angiogenesis and the consequences when this is disrupted can be seen in a variety of congenital and acquired disease states. This review details the mechanisms of vasculogenesis in vivo during embryogenesis and compares this to currently employed in vitro techniques. It also highlights clinical consequences of defects in the endothelial cell-pericyte cross-talk and highlights therapies which are being developed to target this pathway. Improving the understanding of the intricacies of endothelial-pericyte signaling will inform pathophysiology of multiple vascular diseases and allow the development of effective in vitro models to guide drug development and assist with approaches in tissue engineering to develop functional vasculature for regenerative medicine applications.
Collapse
Affiliation(s)
- Mark Sweeney
- Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Gabor Foldes
- Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| |
Collapse
|
100
|
Matsuzaki T, Matsumoto S, Kasai T, Yoshizawa E, Okamoto S, Yoshikawa HY, Taniguchi H, Takebe T. Defining Lineage-Specific Membrane Fluidity Signatures that Regulate Adhesion Kinetics. Stem Cell Reports 2018; 11:852-860. [PMID: 30197117 PMCID: PMC6178887 DOI: 10.1016/j.stemcr.2018.08.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/10/2018] [Accepted: 08/11/2018] [Indexed: 12/13/2022] Open
Abstract
Cellular membrane fluidity is a critical modulator of cell adhesion and migration, prompting us to define the systematic landscape of lineage-specific cellular fluidity throughout differentiation. Here, we have unveiled membrane fluidity landscapes in various lineages ranging from human pluripotency to differentiated progeny: (1) membrane rigidification precedes the exit from pluripotency, (2) membrane composition modulates activin signaling transmission, and (3) signatures are relatively germ layer specific presumably due to unique lipid compositions. By modulating variable lineage-specific fluidity, we developed a label-free “adhesion sorting (AdSort)” method with simple cultural manipulation, effectively eliminating pluripotent stem cells and purifying target population as a result of the over 1,150 of screened conditions combining compounds and matrices. These results underscore the important role of tunable membrane fluidity in influencing stem cell maintenance and differentiation that can be translated into lineage-specific cell purification strategy. Membrane rigidification precedes the exit from pluripotency Germ layer-specific membrane fluidity signature exists Identification of polyphenols as a membrane fluidity modulator Fluidity-based adhesion sorting purify differentiated progeny from pluripotency
Collapse
Affiliation(s)
- Takahisa Matsuzaki
- Institute of Research, Tokyo Medical and Dental University (TMDU), 15-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Shinya Matsumoto
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Toshiharu Kasai
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Emi Yoshizawa
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Satoshi Okamoto
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Hiroshi Y Yoshikawa
- Department of Chemistry, Saitama University, Shimo-okubo 255, Sakura-ku, Saitama 338-8570, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan; Advanced Medical Research Center, Yokohama City University, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Takanori Takebe
- Institute of Research, Tokyo Medical and Dental University (TMDU), 15-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan; Advanced Medical Research Center, Yokohama City University, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan; Division of Gastroenterology, Hepatology & Nutrition, Developmental Biology, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA.
| |
Collapse
|