1
|
Sahajpal NS, Dean J, Hilton B, Fee T, Skinner C, Hastie A, DuPont BR, Chaubey A, Friez MJ, Stevenson RE. Optical genome mapping identifies rare structural variants in neural tube defects. Genome Res 2025; 35:798-809. [PMID: 40107724 PMCID: PMC12047250 DOI: 10.1101/gr.279318.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025]
Abstract
Neural tube defects (NTDs) are the most common birth defects of the central nervous system and occur as either isolated malformations or accompanied by anomalies of other systems. The genetic basis of NTDs remains poorly understood using karyotyping, chromosomal microarray, and short-read sequencing, with only a limited number of pathogenic variants identified. Collectively, these technologies may fail to detect rare structural variants (SVs) in the genome, which may cause these birth defects. Therefore, optical genome mapping (OGM) was applied to investigate 104 NTD cases, of which 74 were isolated NTDs and 30 were NTDs with other malformations. A stepwise approach was undertaken to ascertain candidate variants using population and internal databases and performing parental studies when possible. This analysis identifies diagnostic findings in 8% of cases (8/104) and candidate findings in an additional 22% of cases (23/104). Of the candidate findings, 9% of cases (9/104) have SVs impacting genes associated with NTDs in mouse, and 13% of cases (14/104) have SVs impacting genes implicated in the neural tube development pathways. This study identifies RMND5A, HNRNPC, FOXD4, and RBBP4 as strong candidate genes associated with NTDs, and expands the phenotypic spectrum of AMER1 and TGIF1 to include NTDs. This study constitutes the first systematic investigation of SVs using OGM to elucidate the genetic determinants of NTDs. The data provide key insights into the pathogenesis of NTDs and demonstrate the contribution of SVs in the genome to these birth defects.
Collapse
Affiliation(s)
| | - Jane Dean
- Greenwood Genetic Center, Greenwood, South Carolina 29646, USA
| | - Benjamin Hilton
- Greenwood Genetic Center, Greenwood, South Carolina 29646, USA
| | - Timothy Fee
- Greenwood Genetic Center, Greenwood, South Carolina 29646, USA
| | - Cindy Skinner
- Greenwood Genetic Center, Greenwood, South Carolina 29646, USA
| | - Alex Hastie
- Bionano Genomics, San Diego, California 92121, USA
| | | | - Alka Chaubey
- Bionano Genomics, San Diego, California 92121, USA
| | - Michael J Friez
- Greenwood Genetic Center, Greenwood, South Carolina 29646, USA
| | - Roger E Stevenson
- Greenwood Genetic Center, Greenwood, South Carolina 29646, USA;
- Equanimitas, Greenwood, South Carolina 29646, USA
| |
Collapse
|
2
|
Rybkowska P, Kawalec M, Dymkowska D, Radoszkiewicz K, Zabłocka B, Zabłocki K, Sarnowska A. Activity and function of auxiliary fluxes of glucose metabolism in response to physiological normoxia (5 % O 2) during long-term Adipose-Derived Stem/Stromal cell culture. Eur J Cell Biol 2025; 104:151486. [PMID: 40187000 DOI: 10.1016/j.ejcb.2025.151486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
Energy metabolism homeostasis emerges as a dominant element influencing mesenchymal stem/stromal cells' trajectory of development. The predominant glycolysis activity is a primary driver of cell proliferation and maintenance of the high-energetic state. Here, we examined the functions of two crucial auxiliary pathways: the phosphate-pentose pathway (PPP) and fructose-2,6-biphosphate pathway (FBP) to evaluate their impact on the therapeutic potential of Adipose-Derived Stem/Stromal cells (ASCs) during prolonged culture in various oxygen conditions: 5 % O2 - physiological normoxia or 21 % O2 - atmospheric oxygen. Our findings demonstrate that ASCs cultured in 5 % O2 increased the rate of proliferation, migration, and expression of stemness factors, which is prominent during the initial and middle passages. Additionally, ASCs cultured in a 5 % O2 exhibited heightened protection mechanisms against free radicals, increased LDH gene expression, and elevated extracellular acidification rate (ECAR). By estimating the HIF-1α level, we concluded that 5 % oxygen conditions were insufficient to induce a profound hypoxic state in ASCs. However, at the protein level, both the PPP and FBP pathways appeared to be more active in young (2-passage) cells, regardless of oxygen conditions, and their activity diminished over time. Additionally, the chemical suppression of G6PDH by Polydatin and inhibition of PFKFB3 by PFK-158 in ASCs (passage-2) revealed dose- and time-dependent effect on decreasing migratory capabilities of cells. Nevertheless, our work underscores the adaptable nature of ASC metabolism to prevailing external conditions, with the aging of the culture contributing to the decline in glycolysis-associated auxiliary pathways.
Collapse
Affiliation(s)
- Paulina Rybkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, Warsaw 02-106, Poland.
| | - Maria Kawalec
- Molecular Biology Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, Warsaw 02-106, Poland
| | - Dorota Dymkowska
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3 Street, Warsaw 02-093, Poland
| | - Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, Warsaw 02-106, Poland
| | - Barbara Zabłocka
- Molecular Biology Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, Warsaw 02-106, Poland
| | - Krzysztof Zabłocki
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3 Street, Warsaw 02-093, Poland
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, Warsaw 02-106, Poland.
| |
Collapse
|
3
|
Wang X, Huang W, Sun H, Wang H, Wang D, Wang Y. Tomatidine relieves neuronal damage in spinal cord injury by inhibiting the inflammatory responses and apoptosis through blocking the NF-κB/CXCL10 pathway activation. Front Pharmacol 2024; 15:1503925. [PMID: 39726790 PMCID: PMC11669516 DOI: 10.3389/fphar.2024.1503925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Background Spinal cord injury (SCI) is a neurological disease characterized by high disability and mortality rates. Tomatidine, a natural steroid alkaloid, has been evidenced to have neuroprotective properties. However, the underlying mechanisms of tomatidine in treating SCI remain ambiguous. This study aimed to illustrate the molecular mechanisms of tomatidine in modulating the inflammatory response and promoting functional rehabilitation after SCI. Methods Sprague-Dawley (SD) rats were used to construct an in vivo SCI model and were intraperitoneally injected with tomatidine (5, 10, or 20 mg/kg) for 7 days, followed by treatment with the nuclear factor-κB (NF-κB) pathway agonist (PMA). In addition, lipopolysaccharide (LPS)-induced PC-12 cells were used to establish an SCI cell model and were stimulated with tomatidine, PMA, or a CXCL10 inhibitor. The pathophysiological changes and neurological function were evaluated using blood-brain barrier (BBB) scoring, water content determination, hematoxylin and eosin (H&E) staining, and TUNEL assay. Levels of inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6, were measured. Cell proliferation, apoptosis, and the expression of C-X-C motif chemokine ligand 10 (CXCL10) were determined. Moreover, the expression of cleaved-caspase 3, caspase 3, CXCL10, p-p65, and p65 were analyzed. Results Our data revealed that tomatidine promoted neuronal damage recovery, reduced histopathological changes, elevated cell proliferation, and inhibited the apoptosis and inflammatory factor levels in spinal cord tissues and LPS-induced PC-12 cells. Moreover, tomatidine decreased the expression of CXCL10 in vitro and in vivo, which was accompanied by the regulation of the NF-κB pathway. However, the NF-κB pathway agonist PMA reversed the protective effect of tomatidine in vitro. PMA also enhanced the CXCL10 expression and stimulated the activation of the NF-κB pathway, as demonstrated by the upregulation of phosphorylated p65. The CXCL10 inhibitor had effects similar to tomatidine on cleaved-caspase 3 expression, CXCL10 expression, and the NF-κB pathway. Conclusion Tomatidine can alleviate neuronal damage in SCI by inhibiting apoptosis and inflammation through the NF-κB/CXCL10 pathway. Our findings provide a novel therapeutic target and candidate for the treatment of SCI.
Collapse
Affiliation(s)
- Xu Wang
- The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou, China
- Department of Trauma Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Wei Huang
- Health Management Center, Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Hao Sun
- Department of Trauma Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Hua Wang
- Department of Trauma Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Yongxiang Wang
- The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou, China
- Department of Trauma Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
| |
Collapse
|
4
|
Forston MD, Wei GZ, Chariker JH, Stephenson T, Andres K, Glover C, Rouchka EC, Whittemore SR, Hetman M. Enhanced oxidative phosphorylation, re-organized intracellular signaling, and epigenetic de-silencing as revealed by oligodendrocyte translatome analysis after contusive spinal cord injury. Sci Rep 2023; 13:21254. [PMID: 38040794 PMCID: PMC10692148 DOI: 10.1038/s41598-023-48425-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/27/2023] [Indexed: 12/03/2023] Open
Abstract
Reducing the loss of oligodendrocytes (OLs) is a major goal for neuroprotection after spinal cord injury (SCI). Therefore, the OL translatome was determined in Ribotag:Plp1-CreERT2 mice at 2, 10, and 42 days after moderate contusive T9 SCI. At 2 and 42 days, mitochondrial respiration- or actin cytoskeleton/cell junction/cell adhesion mRNAs were upregulated or downregulated, respectively. The latter effect suggests myelin sheath loss/morphological simplification which is consistent with downregulation of cholesterol biosynthesis transcripts on days 10 and 42. Various regulators of pro-survival-, cell death-, and/or oxidative stress response pathways showed peak expression acutely, on day 2. Many acutely upregulated OL genes are part of the repressive SUZ12/PRC2 operon suggesting that epigenetic de-silencing contributes to SCI effects on OL gene expression. Acute OL upregulation of the iron oxidoreductase Steap3 was confirmed at the protein level and replicated in cultured OLs treated with the mitochondrial uncoupler FCCP. Hence, STEAP3 upregulation may mark mitochondrial dysfunction. Taken together, in SCI-challenged OLs, acute and subchronic enhancement of mitochondrial respiration may be driven by axonal loss and subsequent myelin sheath degeneration. Acutely, the OL switch to oxidative phosphorylation may lead to oxidative stress that is further amplified by upregulation of such enzymes as STEAP3.
Collapse
Affiliation(s)
- Michael D Forston
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - George Z Wei
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- MD/PhD Program, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Julia H Chariker
- Kentucky IDeA Networks of Biomedical Research Excellence (KY INBRE) Bioinformatics Core, University of Louisville, Louisville, KY, 40202, USA
- Neuroscience Training, University Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Tyler Stephenson
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Kariena Andres
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Charles Glover
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Eric C Rouchka
- Kentucky IDeA Networks of Biomedical Research Excellence (KY INBRE) Bioinformatics Core, University of Louisville, Louisville, KY, 40202, USA
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Scott R Whittemore
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- MD/PhD Program, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- MD/PhD Program, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| |
Collapse
|
5
|
Kocher K, Bhattacharya S, Niforatos-Andescavage N, Almalvez M, Henderson D, Vilain E, Limperopoulos C, Délot EC. Genome-wide neonatal epigenetic changes associated with maternal exposure to the COVID-19 pandemic. BMC Med Genomics 2023; 16:268. [PMID: 37899449 PMCID: PMC10614377 DOI: 10.1186/s12920-023-01707-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 10/17/2023] [Indexed: 10/31/2023] Open
Abstract
BACKGROUND During gestation, stressors to the fetus, including viral exposure or maternal psychological distress, can fundamentally alter the neonatal epigenome, and may be associated with long-term impaired developmental outcomes. The impact of in utero exposure to the COVID-19 pandemic on the newborn epigenome has yet to be described. METHODS This study aimed to determine whether there are unique epigenetic signatures in newborns who experienced otherwise healthy pregnancies that occurred during the COVID-19 pandemic (Project RESCUE). The pre-pandemic control and pandemic cohorts (Project RESCUE) included in this study are part of a prospective observational and longitudinal cohort study that evaluates the impact of elevated prenatal maternal stress during the COVID-19 pandemic on early childhood neurodevelopment. Using buccal swabs collected at birth, differential DNA methylation analysis was performed using the Infinium MethylationEPIC arrays and linear regression analysis. Pathway analysis and gene ontology enrichment were performed on resultant gene lists. RESULTS Widespread differential methylation was found between neonates exposed in utero to the pandemic and pre-pandemic neonates. In contrast, there were no apparent epigenetic differences associated with maternal COVID-19 infection during pregnancy. Differential methylation was observed among genomic sites that underpin important neurological pathways that have been previously reported in the literature to be differentially methylated because of prenatal stress, such as NR3C1. CONCLUSIONS The present study reveals potential associations between exposure to the COVID-19 pandemic during pregnancy and subsequent changes in the newborn epigenome. While this finding warrants further investigation, it is a point that should be considered in any study assessing newborn DNA methylation studies obtained during this period, even in otherwise healthy pregnancies.
Collapse
Affiliation(s)
- Kristen Kocher
- Center for Genetic Medicine Research, Children's National Research & Innovation Campus, Washington, DC, USA
- Department of Genomics & Precision Medicine, George Washington University, Washington, DC, USA
| | - Surajit Bhattacharya
- Center for Genetic Medicine Research, Children's National Research & Innovation Campus, Washington, DC, USA
| | | | - Miguel Almalvez
- Institute for Clinical and Translational Science, University of California, Irvine, CA, USA
| | - Diedtra Henderson
- Developing Brain Institute, Children's National Hospital, Washington, DC, USA
| | - Eric Vilain
- Institute for Clinical and Translational Science, University of California, Irvine, CA, USA.
| | | | - Emmanuèle C Délot
- Center for Genetic Medicine Research, Children's National Research & Innovation Campus, Washington, DC, USA.
- Department of Genomics & Precision Medicine, George Washington University, Washington, DC, USA.
| |
Collapse
|
6
|
GÜZEL S, YALÇIN A, GÜRPINAR Y, GÜLER S. Expression of Pfkfb isoenzymes during in vitro differentiation of mouse embryonic stem cells into insulin-producing cells. Turk J Med Sci 2023; 53:1565-1573. [PMID: 38813509 PMCID: PMC10760535 DOI: 10.55730/1300-0144.5725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 12/12/2023] [Accepted: 08/11/2023] [Indexed: 05/31/2024] Open
Abstract
Background/aim Type 1 diabetes mellitus (T1DM) is caused by the autoimmune-mediated destruction of insulin-producing cells (IPCs) and still has no effective cure. Better understanding of the molecular mechanisms involved in the differentiation of embryonic stem cells (ESCs) into IPCs may help us improve the therapeutic strategies for treating T1DM. 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases (Pfkfb1-4) are key regulators of glucose metabolism. Although Pfkfb3 has been shown to be required for the growth of early differentiated mouse ESCs (mESCs), more studies are needed to further assess the roles of Pfkfb isoenzymes in embryonic development and differentiation, particularly into specific cell types. In this study, we aimed to elucidate the changes in the expression of Pfkfb isoenzymes on the differentiation of mESCs into IPCs. Materials and methods A 3-step protocol was used to differentiate R1 and J1 mESCs into IPCs. The changes in the gene expression of MafA, MafB, Ins2, and Nkx6.1 (IPC specific markers) and Pfkfb1-4 were analyzed using real-time quantitative polymerase chain reaction (qPCR). Insulin expression and secretion were determined by immunofluorescence (IF) staining and the enzyme linked immunosorbent assay (ELISA), respectively. Results Upon differentiation, the IPC specific markers in differentiated cells were upregulated. Continued differentiation was confirmed by the development of insulin-positive islet-like clusters that secreted insulin in response to glucose uptake. Expressions of the Pfkfb2 and Pfkfb3 isoenzymes were markedly increased in various stages of differentiation. Conclusion These findings suggest that Pfkfb2 and Pfkfb3 may impact the differentiation of mESCs into IPCs and the regulation of the insulin response to glucose levels. This study also lays a foundation for researchers to further probe the roles of Pfkfb isoenzymes on the differentiation of mESCs into IPCs and may open new avenues for regenerative medicine.
Collapse
Affiliation(s)
- Saime GÜZEL
- Department of Biochemistry, Faculty of Veterinary Medicine, Bursa Uludağ University, Bursa,
Turkiye
| | - Abdullah YALÇIN
- Department of Biochemistry, Faculty of Veterinary Medicine, Bursa Uludağ University, Bursa,
Turkiye
| | - Yunus GÜRPINAR
- Research Center for Translational Medicine, Koç University, İstanbul,
Turkiye
| | - Sabire GÜLER
- Department of Histology & Embryology, Faculty of Veterinary Medicine, Bursa Uludağ University, Bursa,
Turkiye
| |
Collapse
|
7
|
Forston MD, Wei G, Chariker JH, Stephenson T, Andres K, Glover C, Rouchka EC, Whittemore SR, Hetman M. Enhanced oxidative phosphorylation, re-organized intracellular signaling, and epigenetic de-silencing as revealed by oligodendrocyte translatome analysis after contusive spinal cord injury. RESEARCH SQUARE 2023:rs.3.rs-3164618. [PMID: 37546871 PMCID: PMC10402259 DOI: 10.21203/rs.3.rs-3164618/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Reducing the loss of oligodendrocytes (OLs) is a major goal for neuroprotection after spinal cord injury (SCI). Therefore, the OL translatome was determined in Ribotag:Plp1-CreERT2 mice at 2, 10, and 42 days after moderate contusive T9 SCI. At 2 and 42 days, mitochondrial respiration- or actin cytoskeleton/cell junction/cell adhesion mRNAs were upregulated or downregulated, respectively. The latter effect suggests myelin sheath loss/morphological simplification which is consistent with downregulation of cholesterol biosynthesis transcripts on days 10 and 42. Various regulators of pro-survival-, cell death-, and/or oxidative stress response pathways showed peak expression acutely, on day 2. Many acutely upregulated OL genes are part of the repressive SUZ12/PRC2 operon suggesting that epigenetic de-silencing contributes to SCI effects on OL gene expression. Acute OL upregulation of the iron oxidoreductase Steap3 was confirmed at the protein level and replicated in cultured OLs treated with the mitochondrial uncoupler FCCP. Hence, STEAP3 upregulation may mark mitochondrial dysfunction. Taken together, in SCI-challenged OLs, acute and subchronic enhancement of mitochondrial respiration may be driven by axonal loss and subsequent myelin sheath degeneration. Acutely, the OL switch to oxidative phosphorylation may lead to oxidative stress that is further amplified by upregulation of such enzymes as STEAP3.
Collapse
Affiliation(s)
| | - George Wei
- University of Louisville School of Medicine
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Guzel S, Gurpinar Y, Altunok TH, Yalcin A. Increased expression of 6-phosphofructo-2-kinase/fructose 2,6-bisphosphatase-3 is required for growth of mouse embryonic stem cells that are undergoing differentiation. Cytotechnology 2023; 75:27-38. [PMID: 36713065 PMCID: PMC9880118 DOI: 10.1007/s10616-022-00557-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/01/2022] [Indexed: 11/11/2022] Open
Abstract
The unlimited proliferation capacity of embryonic stem cells (ESCs) coupled with their capability to differentiate into several cell types makes them an attractive candidate for studying the molecular mechanisms regulating self-renewal and transition from pluripotent state. Although the roles of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase family (PFKFB1-4) in cell survival, proliferation, and differentiation in tumor cells have been studied, their role in mouse ESC (mESC) biology is currently unkown. In the current study, Pfkfb isoenzyme expressions were analyzed in R1 and J1 mESCs that were cultured in the presence and absence of leukemia inhibitory factor (LIF). We report that expression of the Pfkfb3 isoenzyme was markedly increased when mESCs were promoted to differentiate upon LIF removal. We then demonstrated that Pfkfb3 silencing induced the differentiation marker Brachyury suggesting that Pfkfb3 may be required for the regulation of mesodermal differentiation of mESCs. Furthermore, we show that the increase in Pfkfb3 expression is required for the growth of early differentiated mESCs. Although these results provide important insights into the early differentiation of mESCs with regard to Pfkfb expressions, further mechanistic studies will be needed for understanding the pathways and mechanisms involved in regulation of proliferation and early differentiation of mESCs through Pfkfb3.
Collapse
Affiliation(s)
- Saime Guzel
- Department of Biochemistry, School of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Yunus Gurpinar
- Research Center for Translational Medicine, Koc University, 34010 Istanbul, Turkey
| | - Tugba Hazal Altunok
- Department of Biochemistry, School of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Abdullah Yalcin
- Department of Biochemistry, School of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| |
Collapse
|
9
|
Sittewelle M, Kappès V, Zhou C, Lécuyer D, Monsoro-Burq AH. PFKFB4 interacts with ICMT and activates RAS/AKT signaling-dependent cell migration in melanoma. Life Sci Alliance 2022; 5:5/12/e202201377. [PMID: 35914811 PMCID: PMC9348664 DOI: 10.26508/lsa.202201377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022] Open
Abstract
Glycolysis regulator PFKFB4 promotes cell migration in metastatic melanoma and normal melanocytes by a non-conventional glycolysis-independent function involving ICMT, RAS, and AKT signaling. Cell migration is a complex process, tightly regulated during embryonic development and abnormally activated during cancer metastasis. RAS-dependent signaling is a major nexus controlling essential cell parameters including proliferation, survival, and migration, utilizing downstream effectors such as the PI3K/AKT signaling pathway. In melanoma, oncogenic mutations frequently enhance RAS, PI3K/AKT, or MAP kinase signaling and trigger other cancer hallmarks among which the activation of metabolism regulators. PFKFB4 is one of these critical regulators of glycolysis and of the Warburg effect. Here, however, we explore a novel function of PFKFB4 in melanoma cell migration. We find that PFKFB4 interacts with ICMT, a posttranslational modifier of RAS. PFKFB4 promotes ICMT/RAS interaction, controls RAS localization at the plasma membrane, activates AKT signaling and enhances cell migration. We thus provide evidence of a novel and glycolysis-independent function of PFKFB4 in human cancer cells. This unconventional activity links the metabolic regulator PFKFB4 to RAS-AKT signaling and impacts melanoma cell migration.
Collapse
Affiliation(s)
- Méghane Sittewelle
- Université Paris-Saclay, Faculté des Sciences d'Orsay, CNRS UMR 3347, INSERM U1021, Orsay, France.,Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
| | - Vincent Kappès
- Université Paris-Saclay, Faculté des Sciences d'Orsay, CNRS UMR 3347, INSERM U1021, Orsay, France.,Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
| | - Chenxi Zhou
- Université Paris-Saclay, Faculté des Sciences d'Orsay, CNRS UMR 3347, INSERM U1021, Orsay, France.,Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
| | - Déborah Lécuyer
- Université Paris-Saclay, Faculté des Sciences d'Orsay, CNRS UMR 3347, INSERM U1021, Orsay, France.,Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
| | - Anne H Monsoro-Burq
- Université Paris-Saclay, Faculté des Sciences d'Orsay, CNRS UMR 3347, INSERM U1021, Orsay, France .,Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
| |
Collapse
|
10
|
Yang H, Shao N, Holmström A, Zhao X, Chour T, Chen H, Itzhaki I, Wu H, Ameen M, Cunningham NJ, Tu C, Zhao MT, Tarantal AF, Abilez OJ, Wu JC. Transcriptome analysis of non human primate-induced pluripotent stem cell-derived cardiomyocytes in 2D monolayer culture vs. 3D engineered heart tissue. Cardiovasc Res 2021; 117:2125-2136. [PMID: 33002105 PMCID: PMC8318103 DOI: 10.1093/cvr/cvaa281] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/27/2020] [Accepted: 09/17/2020] [Indexed: 12/22/2022] Open
Abstract
AIMS Stem cell therapy has shown promise for treating myocardial infarction via re-muscularization and paracrine signalling in both small and large animals. Non-human primates (NHPs), such as rhesus macaques (Macaca mulatta), are primarily utilized in preclinical trials due to their similarity to humans, both genetically and physiologically. Currently, induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) are delivered into the infarcted myocardium by either direct cell injection or an engineered tissue patch. Although both approaches have advantages in terms of sample preparation, cell-host interaction, and engraftment, how the iPSC-CMs respond to ischaemic conditions in the infarcted heart under these two different delivery approaches remains unclear. Here, we aim to gain a better understanding of the effects of hypoxia on iPSC-CMs at the transcriptome level. METHODS AND RESULTS NHP iPSC-CMs in both monolayer culture (2D) and engineered heart tissue (EHT) (3D) format were exposed to hypoxic conditions to serve as surrogates of direct cell injection and tissue implantation in vivo, respectively. Outcomes were compared at the transcriptome level. We found the 3D EHT model was more sensitive to ischaemic conditions and similar to the native in vivo myocardium in terms of cell-extracellular matrix/cell-cell interactions, energy metabolism, and paracrine signalling. CONCLUSION By exposing NHP iPSC-CMs to different culture conditions, transcriptome profiling improves our understanding of the mechanism of ischaemic injury.
Collapse
Affiliation(s)
- Huaxiao Yang
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Division of Cardiology, Department of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Department of Biomedical Engineering, University of North Texas, 390 N. Elm Street K240B, Denton, TX 76207-7102, USA
| | - Ningyi Shao
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Division of Cardiology, Department of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
| | - Alexandra Holmström
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Division of Cardiology, Department of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
| | - Xin Zhao
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Division of Cardiology, Department of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
| | - Tony Chour
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Division of Cardiology, Department of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
| | - Haodong Chen
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Division of Cardiology, Department of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
| | - Ilanit Itzhaki
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Division of Cardiology, Department of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
| | - Haodi Wu
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Division of Cardiology, Department of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
| | - Mohamed Ameen
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Division of Cardiology, Department of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
| | - Nathan J Cunningham
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Division of Cardiology, Department of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
| | - Chengyi Tu
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Division of Cardiology, Department of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
| | - Ming-Tao Zhao
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Division of Cardiology, Department of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
| | - Alice F Tarantal
- Department of Pediatrics, School of Medicine, One Shields Avenue, Davis, CA 95616-8542, USA
- Department Cell Biology and Human Anatomy, School of Medicine, One Shields Avenue, Davis, CA 95616-8542, USA
- California National Primate Research Center, UC Davis, One Shields Avenue, Davis, CA 95616-8542, USA
| | - Oscar J Abilez
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Division of Cardiology, Department of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Division of Cardiology, Department of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive G1120B, Stanford, CA 94305-5454, USA
| |
Collapse
|
11
|
Alkobtawi M, Pla P, Monsoro-Burq AH. BMP signaling is enhanced intracellularly by FHL3 controlling WNT-dependent spatiotemporal emergence of the neural crest. Cell Rep 2021; 35:109289. [PMID: 34161771 DOI: 10.1016/j.celrep.2021.109289] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/23/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023] Open
Abstract
The spatiotemporal coordination of multiple morphogens is essential for embryonic patterning yet poorly understood. During neural crest (NC) formation, dynamic bone morphogenetic protein (BMP), fibroblast growth factor (FGF), and WNT signals cooperate by acting on mesoderm and ectoderm. Here, we show that Fhl3, a scaffold LIM domain protein, modulates BMP gradient interpretation during NC induction. During gastrulation, low BMP signaling neuralizes the neural border (NB) ectoderm, while Fhl3 enhances Smad1 intracellular response in underlying paraxial mesoderm, triggering the high WNT8 signals needed to pattern the NB. During neurulation, fhl3 activation in NC ectoderm promotes simultaneous high BMP and BMP-dependent WNT activity required for specification. Mechanistically, Fhl3 interacts with Smad1 and promotes Smad1 binding to wnt8 promoter in a BMP-dependent manner. Consequently, differential Fhl3 expression in adjacent cells ensures a finely tuned coordination of BMP and WNT signaling at several stages of NC development, starting by positioning the NC-inducing mesoderm center under competent NB ectoderm.
Collapse
Affiliation(s)
- Mansour Alkobtawi
- Université Paris-Saclay, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, rue Henri Becquerel, F-91405 Orsay, France
| | - Patrick Pla
- Université Paris-Saclay, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, rue Henri Becquerel, F-91405 Orsay, France
| | - Anne H Monsoro-Burq
- Université Paris-Saclay, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, rue Henri Becquerel, F-91405 Orsay, France; Institut Universitaire de France, F-75005 Paris, France.
| |
Collapse
|
12
|
PFKFB4 Overexpression Facilitates Proliferation by Promoting the G1/S Transition and Is Associated with a Poor Prognosis in Triple-Negative Breast Cancer. DISEASE MARKERS 2021; 2021:8824589. [PMID: 34211613 PMCID: PMC8211511 DOI: 10.1155/2021/8824589] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 02/12/2021] [Accepted: 04/12/2021] [Indexed: 12/31/2022]
Abstract
Background 6-Phosphofructo-2-kinase/fructose-2,6-biphosphate-4 (PFKFB4) is a key factor that plays an important role in tumorigenesis. However, its role in triple-negative breast cancer (TNBC) progression needs to be further validated. We investigated whether PFKFB4 is directly involved in the oncogenic signaling networks of TNBC. Methods First, we assessed the expression level of PFKFB4 in tumor tissue specimens by immunohistochemistry and evaluated its prognostic value. Next, the effect of PFKFB4 on TNBC cell growth and associated mechanisms were investigated. Finally, the results were further verified in vivo. Results We found that PFKFB4 overexpression was associated with an unfavorable prognosis in TNBC patients. PFKFB4 was overexpressed in TNBC cell lines in hypoxic environments, and its overexpression promoted tumor progression in vitro and in vivo. Further analyses demonstrated that the possible mechanism might be that PFKFB4 overexpression facilitates TNBC progression by enhancing the G1/S phase transition by increasing the protein level of CDK6 and phosphorylation of Rb. Conclusions These data suggest that PFKFB4 plays significant roles in the tumorigenesis and development of TNBC.
Collapse
|
13
|
Zhang L, Liu Z, Dong Y, Kong L. E2F2 drives glioma progression via PI3K/AKT in a PFKFB4-dependent manner. Life Sci 2021; 276:119412. [PMID: 33774025 DOI: 10.1016/j.lfs.2021.119412] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
AIMS The effects of PFKFB4 on glycolysis during the cancer progression has been investigated, while its role in glioma remains unclear. The present study evaluated the molecular mechanism of PFKFB4 in glycolysis of glioma progression. MATERIALS AND METHODS The pan-cancer platform SangerBox was inquired to investigate the E2F2 expression in tumors. The E2F2 expression was studied by qRT-PCR and immunohistochemistry in collected glioma and normal brain tissues and by qRT-PCR and western blot in glioma cells. The relationship between the E2F2 expression in glioma tissues and patients' prognosis was analyzed. The cell malignant phenotype, glycolysis, growth and metastasis were examined by CCK-8, EdU, colony formation, flow cytometry, wound healing, Transwell assays, ELISA kits, and tumorigenesis and metastasis assays. Downstream targets of E2F2 were searched in hTFtarget, followed by pathway enrichment analysis. The expression of these targets and their correlation with E2F2 expression in gliomas were investigated through the GEPIA website. After ChIP and luciferase assays, the effect of the target on glioma was investigated. KEY FINDINGS E2F2 was overexpressed in glioma patients and predicted poor prognoses. E2F2 promoted cell proliferation, colony formation, DNA synthesis, migration, invasion and glycolysis, and inhibited apoptosis. Meanwhile, inhibition of E2F2 suppressed the growth and metastasis of gliomas. E2F2 elevated the PFKFB4 expression transcriptionally by binding to its promoter and activated PI3K/AKT pathway. The promotion of glioma metastasis and glycolysis by E2F2 was mitigated by PFKFB4 knockdown. SIGNIFICANCE E2F2-mediated transcriptional enhancement of PFKFB4 expression regulated the phosphorylation of PI3K/AKT to promote glioma malignancy progression.
Collapse
Affiliation(s)
- Longzhou Zhang
- Department of Neurosurgery, First Hospital Affiliated to Zhengzhou University, Zhengzhou 450000, Henan, PR China.
| | - Zengjin Liu
- Department of Neurosurgery, First Hospital Affiliated to Zhengzhou University, Zhengzhou 450000, Henan, PR China
| | - Yang Dong
- Department of Neurosurgery, First Hospital Affiliated to Zhengzhou University, Zhengzhou 450000, Henan, PR China
| | - Lingchang Kong
- Department of Neurosurgery, ZhengZhou Traditional Chinese Medicine Hospital, Zhengzhou 450000, Henan, PR China
| |
Collapse
|
14
|
Perfetto M, Xu X, Lu C, Shi Y, Yousaf N, Li J, Yien YY, Wei S. The RNA helicase DDX3 induces neural crest by promoting AKT activity. Development 2021; 148:dev.184341. [PMID: 33318149 DOI: 10.1242/dev.184341] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/02/2020] [Indexed: 01/02/2023]
Abstract
Mutations in the RNA helicase DDX3 have emerged as a frequent cause of intellectual disability in humans. Because many individuals carrying DDX3 mutations have additional defects in craniofacial structures and other tissues containing neural crest (NC)-derived cells, we hypothesized that DDX3 is also important for NC development. Using Xenopus tropicalis as a model, we show that DDX3 is required for normal NC induction and craniofacial morphogenesis by regulating AKT kinase activity. Depletion of DDX3 decreases AKT activity and AKT-dependent inhibitory phosphorylation of GSK3β, leading to reduced levels of β-catenin and Snai1: two GSK3β substrates that are crucial for NC induction. DDX3 function in regulating these downstream signaling events during NC induction is likely mediated by RAC1, a small GTPase whose translation depends on the RNA helicase activity of DDX3. These results suggest an evolutionarily conserved role of DDX3 in NC development by promoting AKT activity, and provide a potential mechanism for the NC-related birth defects displayed by individuals harboring mutations in DDX3 and its downstream effectors in this signaling cascade.
Collapse
Affiliation(s)
- Mark Perfetto
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA.,Department of Biology, West Virginia University, Morgantown, WV 26506, USA
| | - Xiaolu Xu
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Congyu Lu
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Yu Shi
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Natasha Yousaf
- Department of Biology, West Virginia University, Morgantown, WV 26506, USA
| | - Jiejing Li
- Department of Biology, West Virginia University, Morgantown, WV 26506, USA.,Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming 650032, China
| | - Yvette Y Yien
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Shuo Wei
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
15
|
Dong Q, Cheng LY. Getting in shape: ATP pumps up the volume in Hh signalling. EMBO J 2020; 39:e106564. [PMID: 33021741 DOI: 10.15252/embj.2020106564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Glycolysis is a major metabolic process which ensures the break down of glucose into pyruvate via multiple enzymatic steps, but if and how this catabolism can impact on developmental patterning is unclear. In this issue, Spannl et al (2020) demonstrate a novel link between energy metabolism and tissue formation in the fly imaginal discs. They show that ATPs generated via glycolysis maintain active transport of a smoothened inhibitor, which keeps Hh signalling in check to preserve the correct shape and proportion of the developing wing.
Collapse
Affiliation(s)
- Qian Dong
- Peter MacCallum Cancer Institute, Melbourne, Vic., Australia
| | - Louise Y Cheng
- Peter MacCallum Cancer Institute, Melbourne, Vic., Australia
| |
Collapse
|
16
|
Isorhamnetin Induces Melanoma Cell Apoptosis via the PI3K/Akt and NF- κB Pathways. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1057943. [PMID: 32461960 PMCID: PMC7225865 DOI: 10.1155/2020/1057943] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/31/2020] [Indexed: 01/04/2023]
Abstract
Malignant melanoma is characterized by its bad prognosis for aggressiveness, drug resistance, and early metastasis. Isorhamnetin (3′-methoxy-3,4′,5,7-tetrahydroxyflavone; IH) is a natural flavonoid that has been investigated for its antitumor effects in breast cancer, colon cancer, and gastric cancer through inducing cell apoptosis. Given its role in tumor inhibition, no research has been conducted concerning its effect against melanoma. In the present study, we found that IH could significantly inhibit B16F10 cell proliferation and migration and induce B16F10 cell apoptosis. The examination on molecular mechanism revealed that IH could suppress the phosphorylation of Akt and the translocation of NF-κB, which are key factors in apoptosis-related pathways. We also detected that this process was related to the bifunctional 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases 4 (PFKFB4) by PFKFB4 knockdown experiment. In line with in vitro study, we further provided that IH effectively inhibited tumor growth in vivo. Taken together, IH was proven to induce melanoma cell apoptosis in vitro and in vivo, which may serve as a potential agent in malignant melanoma treatment in the future.
Collapse
|
17
|
Prasad MS, Uribe-Querol E, Marquez J, Vadasz S, Yardley N, Shelar PB, Charney RM, García-Castro MI. Blastula stage specification of avian neural crest. Dev Biol 2020; 458:64-74. [PMID: 31610145 PMCID: PMC7050198 DOI: 10.1016/j.ydbio.2019.10.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/09/2019] [Accepted: 10/09/2019] [Indexed: 11/21/2022]
Abstract
Cell fate specification defines the earliest steps towards a distinct cell lineage. Neural crest, a multipotent stem cell population, is thought to be specified from the ectoderm, but its varied contributions defy canons of segregation potential and challenges its embryonic origin. Aiming to resolve this conflict, we have assayed the earliest specification of neural crest using blastula stage chick embryos. Specification assays on isolated chick epiblast explants identify an intermediate region specified towards the neural crest cell fate. Furthermore, low density culture suggests that the specification of intermediate cells towards the neural crest lineage is independent of contact mediated induction and Wnt-ligand induced signaling, but is, however, dependent on transcriptional activity of β-catenin. Finally, we have validated the regional identity of the intermediate region towards the neural crest cell fate using fate map studies. Our results suggest a model of neural crest specification within a restricted epiblast region in blastula stage chick embryos.
Collapse
Affiliation(s)
- Maneeshi S Prasad
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, USA
| | | | | | | | | | - Patrick B Shelar
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, USA
| | - Rebekah M Charney
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, USA
| | - Martín I García-Castro
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, USA.
| |
Collapse
|
18
|
Vanherwegen AS, Eelen G, Ferreira GB, Ghesquière B, Cook DP, Nikolic T, Roep B, Carmeliet P, Telang S, Mathieu C, Gysemans C. Vitamin D controls the capacity of human dendritic cells to induce functional regulatory T cells by regulation of glucose metabolism. J Steroid Biochem Mol Biol 2019; 187:134-145. [PMID: 30481575 DOI: 10.1016/j.jsbmb.2018.11.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/21/2018] [Accepted: 11/23/2018] [Indexed: 12/13/2022]
Abstract
Tolerogenic dendritic cells (tolDCs) instruct regulatory T cells (Tregs) to dampen autoimmunity. Active vitamin D3 (1α,25-dihydroxyvitamin D3; 1α,25(OH)2D3) imprints human monocyte-derived DCs with tolerogenic properties by reprogramming their glucose metabolism. Here we identify the glycolytic enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 (PFKFB4) as a critical checkpoint and direct transcriptional target of 1α,25(OH)2D3 in determining the tolDC profile. Using tracer metabolomics, we show that PFKFB4 activity is essential for glucose metabolism, especially for glucose oxidation, which is elevated upon 1α,25(OH)2D3 exposure. Pharmacological inhibition of PFKFB4 reversed the 1α,25(OH)2D3-mediated shift in metabolism, DC profile and function, as determined by expression of inhibitory surface markers and secretion of regulatory cytokines and factors. Moreover, PFKFB4 inhibition in 1α,25(OH)2D3-treated DCs blocked their hallmark capacity to induce suppressive Tregs. This work demonstrates that alterations in the bioenergetic metabolism of immune cells are central to the immunomodulatory effects induced by 1α,25(OH)2D3.
Collapse
Affiliation(s)
| | - Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Bart Ghesquière
- Metabolomics Core Facility, Center for Cancer Biology, VIB, Leuven,Belgium; Metabolomics Core Facility, Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Tanja Nikolic
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Bart Roep
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands; Department of Diabetes Immunology, Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sucheta Telang
- Division of Hematology/Oncology, Department of Medicine, J. Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, KU, Leuven, Belgium
| | - Conny Gysemans
- Clinical and Experimental Endocrinology, KU, Leuven, Belgium.
| |
Collapse
|
19
|
Tseropoulos G, Moghadasi Boroujeni S, Bajpai VK, Lei P, Andreadis ST. Derivation of neural crest stem cells from human epidermal keratinocytes requires FGF-2, IGF-1, and inhibition of TGF-β1. Bioeng Transl Med 2018; 3:256-264. [PMID: 30377664 PMCID: PMC6195909 DOI: 10.1002/btm2.10109] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/30/2018] [Accepted: 07/30/2018] [Indexed: 11/29/2022] Open
Abstract
Neural crest (NC) cells play a central role in forming the peripheral nervous system, the craniofacial skeleton, and the pigmentation of the skin during development due to their broad multilineage differentiation potential into neurons, Schwann cells, melanocytes, and mesenchymal stem cells. Recently, we identified an easily accessible source of pluripotent NC stem cells from human inter‐follicular keratinocyte (KC) cultures (KC‐NC). In this work, we examined specific conditions for the derivation of NC from KC cultures. More specifically, we examined the role of two growth factors, FGF2 and IGF1, in NC proliferation and in expression of two potent NC transcription factors, Sox10 and FoxD3. Using specific chemical inhibitors, we uncovered that the downstream regulatory pathways AKT/PI3K, MEK/ERK, and JNK/cJun may be critical in Sox10 and FoxD3 regulation in KC‐NC. The TGF‐β1 pathway was also implicated in suppressing Sox10 expression and NC proliferation. In summary, our study shed light into the role of FGF2, IGF1, and TGF‐β1 on the induction of NC from KC cultures and the pathways that regulate Sox10 and FoxD3. We also established culture conditions for sustaining KC‐NC multipotency and, therefore, the potential of these cells for regenerative medicine and cellular therapies.
Collapse
Affiliation(s)
- Georgios Tseropoulos
- Dept. of Chemical and Biological Engineering University at Buffalo Buffalo NY 14260
| | | | - Vivek K Bajpai
- Dept. of Chemical and Biological Engineering University at Buffalo Buffalo NY 14260
| | - Pedro Lei
- Dept. of Chemical and Biological Engineering University at Buffalo Buffalo NY 14260
| | - Stelios T Andreadis
- Dept. of Chemical and Biological Engineering University at Buffalo Buffalo NY 14260.,Dept. of Biomedical Engineering University at Buffalo Buffalo NY 14228.,Center of Excellence in Bioinformatics and Life Sciences Buffalo NY 14203
| |
Collapse
|
20
|
Miyazawa H, Aulehla A. Revisiting the role of metabolism during development. Development 2018; 145:145/19/dev131110. [DOI: 10.1242/dev.131110] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
ABSTRACT
An emerging view emphasizes that metabolism is highly regulated in both time and space. In addition, it is increasingly being recognized that metabolic pathways are tightly connected to specific biological processes such as cell signaling, proliferation and differentiation. As we obtain a better view of this spatiotemporal regulation of metabolism, and of the molecular mechanisms that connect metabolism and signaling, we can now move from largely correlative to more functional studies. It is, therefore, a particularly promising time to revisit how metabolism can affect multiple aspects of animal development. In this Review, we discuss how metabolism is mechanistically linked to cellular and developmental programs through both its bioenergetic and metabolic signaling functions. We highlight how metabolism is regulated across various spatial and temporal scales, and discuss how this regulation can influence cellular processes such as cell signaling, gene expression, and epigenetic and post-translational modifications during embryonic development.
Collapse
Affiliation(s)
- Hidenobu Miyazawa
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, 69117, Germany
| | - Alexander Aulehla
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, 69117, Germany
| |
Collapse
|
21
|
Dai J, Zhou Q, Chen J, Rexius-Hall ML, Rehman J, Zhou G. Alpha-enolase regulates the malignant phenotype of pulmonary artery smooth muscle cells via the AMPK-Akt pathway. Nat Commun 2018; 9:3850. [PMID: 30242159 PMCID: PMC6155017 DOI: 10.1038/s41467-018-06376-x] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 09/03/2018] [Indexed: 12/20/2022] Open
Abstract
The molecular mechanisms underlying the metabolic shift toward increased glycolysis observed in pulmonary artery smooth muscle cells (PASMC) during the pathogenesis of pulmonary arterial hypertension (PAH) are not fully understood. Here we show that the glycolytic enzyme α-enolase (ENO1) regulates the metabolic reprogramming and malignant phenotype of PASMC. We show that ENO1 levels are elevated in patients with associated PAH and in animal models of hypoxic pulmonary hypertension (HPH). The silencing or inhibition of ENO1 decreases PASMC proliferation and de-differentiation, and induces PASMC apoptosis, whereas the overexpression of ENO1 promotes a synthetic, de- differentiated, and apoptotic-resistant phenotype via the AMPK-Akt pathway. The suppression of ENO1 prevents the hypoxia-induced metabolic shift from mitochondrial respiration to glycolysis in PASMC. Finally, we find that pharmacological inhibition of ENO1 reverses HPH in mice and rats, suggesting ENO1 as a regulator of pathogenic metabolic reprogramming in HPH.
Collapse
Affiliation(s)
- Jingbo Dai
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Qiyuan Zhou
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Jiwang Chen
- Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Megan L Rexius-Hall
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Jalees Rehman
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60612, USA
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Guofei Zhou
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, 60612, USA.
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China.
| |
Collapse
|
22
|
Sittewelle M, Monsoro-Burq AH. AKT signaling displays multifaceted functions in neural crest development. Dev Biol 2018; 444 Suppl 1:S144-S155. [PMID: 29859890 DOI: 10.1016/j.ydbio.2018.05.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/24/2018] [Accepted: 05/29/2018] [Indexed: 12/23/2022]
Abstract
AKT signaling is an essential intracellular pathway controlling cell homeostasis, cell proliferation and survival, as well as cell migration and differentiation in adults. Alterations impacting the AKT pathway are involved in many pathological conditions in human disease. Similarly, during development, multiple transmembrane molecules, such as FGF receptors, PDGF receptors or integrins, activate AKT to control embryonic cell proliferation, migration, differentiation, and also cell fate decisions. While many studies in mouse embryos have clearly implicated AKT signaling in the differentiation of several neural crest derivatives, information on AKT functions during the earliest steps of neural crest development had remained relatively scarce until recently. However, recent studies on known and novel regulators of AKT signaling demonstrate that this pathway plays critical roles throughout the development of neural crest progenitors. Non-mammalian models such as fish and frog embryos have been instrumental to our understanding of AKT functions in neural crest development, both in neural crest progenitors and in the neighboring tissues. This review combines current knowledge acquired from all these different vertebrate animal models to describe the various roles of AKT signaling related to neural crest development in vivo. We first describe the importance of AKT signaling in patterning the tissues involved in neural crest induction, namely the dorsal mesoderm and the ectoderm. We then focus on AKT signaling functions in neural crest migration and differentiation.
Collapse
Affiliation(s)
- Méghane Sittewelle
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France
| | - Anne H Monsoro-Burq
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France; Institut Universitaire de France, F-75005 Paris, France.
| |
Collapse
|
23
|
Gándara L, Wappner P. Metabo-Devo: A metabolic perspective of development. Mech Dev 2018; 154:12-23. [PMID: 29475040 DOI: 10.1016/j.mod.2018.02.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/19/2018] [Accepted: 02/19/2018] [Indexed: 02/07/2023]
Abstract
In the last years, several reports have established the notion that metabolism is not just a housekeeping process, but instead an active effector of physiological changes. The idea that the metabolic status may rule a wide range of phenomena in cell biology is starting to be broadly accepted. Thus, current developmental biology has begun to describe different ways by which the metabolic profile of the cell and developmental programs of the organism can crosstalk. In this review, we discuss mechanisms by which metabolism impacts on processes governing development. We review the growing body of evidence that supports the notion that aerobic glycolysis is required in cells undergoing fast growth and high proliferation, similarly to the Warburg effect described in tumor cells. Glycolytic metabolism explains not only the higher ATP synthesis rate required for cell growth, but also the uncoupling between mitochondrial activity and bioenergetics needed to provide anabolism with sufficient precursors. We also discuss some recent studies, which show that in addition to its role in providing energy and carbon chains, the metabolic status of the cell can also influence epigenetic regulation of developmental processes. Although metabolic aspects of development are just starting to be explored, there is no doubt that ongoing research in this field will shape the future landscape of Developmental Biology.
Collapse
Affiliation(s)
- Lautaro Gándara
- Instituto Leloir, Av. Patricias Argentinas 435, Ciudad de Buenos Aires C1405BWE, Argentina
| | - Pablo Wappner
- Instituto Leloir, Av. Patricias Argentinas 435, Ciudad de Buenos Aires C1405BWE, Argentina; Departamento de Fisiología, Biología Molecular, y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
24
|
Geary L, LaBonne C. FGF mediated MAPK and PI3K/Akt Signals make distinct contributions to pluripotency and the establishment of Neural Crest. eLife 2018; 7:33845. [PMID: 29350613 PMCID: PMC5790379 DOI: 10.7554/elife.33845] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/15/2018] [Indexed: 12/12/2022] Open
Abstract
Early vertebrate embryos possess cells with the potential to generate all embryonic cell types. While this pluripotency is progressively lost as cells become lineage restricted, Neural Crest cells retain broad developmental potential. Here, we provide novel insights into signals essential for both pluripotency and neural crest formation in Xenopus. We show that FGF signaling controls a subset of genes expressed by pluripotent blastula cells, and find a striking switch in the signaling cascades activated by FGF signaling as cells lose pluripotency and commence lineage restriction. Pluripotent cells display and require Map Kinase signaling, whereas PI3 Kinase/Akt signals increase as developmental potential is restricted, and are required for transit to certain lineage restricted states. Importantly, retaining a high Map Kinase/low Akt signaling profile is essential for establishing Neural Crest stem cells. These findings shed important light on the signal-mediated control of pluripotency and the molecular mechanisms governing genesis of Neural Crest.
Collapse
Affiliation(s)
- Lauren Geary
- Department of Molecular Biosciences, Northwestern University, Evanston, United States
| | - Carole LaBonne
- Department of Molecular Biosciences, Northwestern University, Evanston, United States.,Robert H Lurie Comprehensive Cancer Center, Northwestern University, Evanston, United States
| |
Collapse
|
25
|
Cowan JR, Tariq M, Shaw C, Rao M, Belmont JW, Lalani SR, Smolarek TA, Ware SM. Copy number variation as a genetic basis for heterotaxy and heterotaxy-spectrum congenital heart defects. Philos Trans R Soc Lond B Biol Sci 2017; 371:rstb.2015.0406. [PMID: 27821535 DOI: 10.1098/rstb.2015.0406] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2016] [Indexed: 12/22/2022] Open
Abstract
Genomic disorders and rare copy number abnormalities are identified in 15-25% of patients with syndromic conditions, but their prevalence in individuals with isolated birth defects is less clear. A spectrum of congenital heart defects (CHDs) is seen in heterotaxy, a highly heritable and genetically heterogeneous multiple congenital anomaly syndrome resulting from failure to properly establish left-right (L-R) organ asymmetry during early embryonic development. To identify novel genetic causes of heterotaxy, we analysed copy number variants (CNVs) in 225 patients with heterotaxy and heterotaxy-spectrum CHDs using array-based genotyping methods. Clinically relevant CNVs were identified in approximately 20% of patients and encompassed both known and putative heterotaxy genes. Patients were carefully phenotyped, revealing a significant association of abdominal situs inversus with pathogenic or likely pathogenic CNVs, while d-transposition of the great arteries was more frequently associated with common CNVs. Identified cytogenetic abnormalities ranged from large unbalanced translocations to smaller, kilobase-scale CNVs, including a rare, single exon deletion in ZIC3, a gene known to cause X-linked heterotaxy. Morpholino loss-of-function experiments in Xenopus support a role for one of these novel candidates, the platelet isoform of phosphofructokinase-1 (PFKP) in heterotaxy. Collectively, our results confirm a high CNV yield for array-based testing in patients with heterotaxy, and support use of CNV analysis for identification of novel biological processes relevant to human laterality.This article is part of the themed issue 'Provocative questions in left-right asymmetry'.
Collapse
Affiliation(s)
- Jason R Cowan
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.,Department of Pediatrics and Medical and Molecular Genetics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Muhammad Tariq
- Department of Pediatrics and Medical and Molecular Genetics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Department of Clinical Biochemistry, University of Tabuk, Tabuk 71491, Kingdom of Saudi Arabia
| | - Chad Shaw
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mitchell Rao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - John W Belmont
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Seema R Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Teresa A Smolarek
- Cincinnati Children's Hospital Medical Center, Division of Human Genetics, Cincinnati, OH 45229, USA
| | - Stephanie M Ware
- Department of Pediatrics and Medical and Molecular Genetics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
26
|
Plouhinec JL, Medina-Ruiz S, Borday C, Bernard E, Vert JP, Eisen MB, Harland RM, Monsoro-Burq AH. A molecular atlas of the developing ectoderm defines neural, neural crest, placode, and nonneural progenitor identity in vertebrates. PLoS Biol 2017; 15:e2004045. [PMID: 29049289 PMCID: PMC5663519 DOI: 10.1371/journal.pbio.2004045] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 10/31/2017] [Accepted: 09/29/2017] [Indexed: 12/18/2022] Open
Abstract
During vertebrate neurulation, the embryonic ectoderm is patterned into lineage progenitors for neural plate, neural crest, placodes and epidermis. Here, we use Xenopus laevis embryos to analyze the spatial and temporal transcriptome of distinct ectodermal domains in the course of neurulation, during the establishment of cell lineages. In order to define the transcriptome of small groups of cells from a single germ layer and to retain spatial information, dorsal and ventral ectoderm was subdivided along the anterior-posterior and medial-lateral axes by microdissections. Principal component analysis on the transcriptomes of these ectoderm fragments primarily identifies embryonic axes and temporal dynamics. This provides a genetic code to define positional information of any ectoderm sample along the anterior-posterior and dorsal-ventral axes directly from its transcriptome. In parallel, we use nonnegative matrix factorization to predict enhanced gene expression maps onto early and mid-neurula embryos, and specific signatures for each ectoderm area. The clustering of spatial and temporal datasets allowed detection of multiple biologically relevant groups (e.g., Wnt signaling, neural crest development, sensory placode specification, ciliogenesis, germ layer specification). We provide an interactive network interface, EctoMap, for exploring synexpression relationships among genes expressed in the neurula, and suggest several strategies to use this comprehensive dataset to address questions in developmental biology as well as stem cell or cancer research. Vertebrate embryo germ layers become progressively regionalized by evolutionarily conserved molecular processes. Catching the early steps of this dynamic spatial cell diversification at the scale of the transcriptome was challenging, even with the advent of efficient RNA sequencing. We have microdissected complementary and defined areas of a single germ layer, the developing ectoderm, and explored how the transcriptome changes over time and space in the ectoderm during the differentiation of frog epidermis, neural plate, and neural crest. We have created EctoMap, a searchable interface using these regional transcriptomes, to predict the expression of the 31 thousand genes expressed in neurulae and their networks of co-expression, predictive of functional relationships. Through several examples, we illustrate how these data provide insights in development, cancer, evolution and stem cell biology.
Collapse
Affiliation(s)
- Jean-Louis Plouhinec
- Université Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Orsay, France
- Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
- MINES ParisTech, PSL Research University, CBIO - Centre for Computational Biology, Paris, France
| | - Sofía Medina-Ruiz
- Department of Molecular and Cell Biology, Division of Genetics, Genomics and Development Biology, University of California, Berkeley, Berkeley, California, United States of America
| | - Caroline Borday
- Université Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Orsay, France
- Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
| | - Elsa Bernard
- MINES ParisTech, PSL Research University, CBIO - Centre for Computational Biology, Paris, France
- Institut Curie, INSERM U900, Paris, France
- INSERM U900, Paris, France
| | - Jean-Philippe Vert
- MINES ParisTech, PSL Research University, CBIO - Centre for Computational Biology, Paris, France
- Institut Curie, INSERM U900, Paris, France
- INSERM U900, Paris, France
| | - Michael B. Eisen
- Department of Molecular and Cell Biology, Division of Genetics, Genomics and Development Biology, University of California, Berkeley, Berkeley, California, United States of America
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, California, United States of America
| | - Richard M. Harland
- Department of Molecular and Cell Biology, Division of Genetics, Genomics and Development Biology, University of California, Berkeley, Berkeley, California, United States of America
| | - Anne H. Monsoro-Burq
- Université Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Orsay, France
- Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
- Institut Universitaire de France, Paris, France
- * E-mail:
| |
Collapse
|
27
|
Taylor C, Mannion D, Miranda F, Karaminejadranjbar M, Herrero-Gonzalez S, Hellner K, Zheng Y, Bartholomeusz G, Bast RC, Ahmed AA. Loss of PFKFB4 induces cell death in mitotically arrested ovarian cancer cells. Oncotarget 2017; 8:17960-17980. [PMID: 28152500 PMCID: PMC5392300 DOI: 10.18632/oncotarget.14910] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 12/15/2016] [Indexed: 12/26/2022] Open
Abstract
Taxanes represent some of the most commonly used chemotherapeutic agents for ovarian cancer treatment. However, they are only effective in approximately 40% of patients. Novel therapeutic strategies are required to potentiate their effect and improve patient outcome. A hallmark of many cancers is the constitutive activation of the PI3K/AKT pathway, which drives cell survival and metabolism. We discovered a striking decrease in AKT activity coupled with a significant reduction in glucose 6-phosphate and ATP levels during mitotic arrest in the majority of ovarian cancer cell lines tested, indicating a potential metabolic vulnerability. A high-content siRNA screen to detect novel metabolic targets in mitotically arrested ovarian cancer cells identified the glycolytic enzyme PFKFB4. PFKFB4 depletion increased caspase 3/7 activity, and levels of reactive oxygen species only in mitotically arrested cells, and significantly enhanced mitotic cell death after paclitaxel treatment. Depletion of PFKFB3 demonstrated a similar phenotype. The observation that some ovarian cancer cells lose AKT activity during mitotic arrest and become vulnerable to metabolic targeting is a new concept in cancer therapy. Thus, combining mitotic-targeted therapies with glycolytic inhibitors may act to potentiate the effects of antimitotics in ovarian cancer through mitosis-specific cell death.
Collapse
Affiliation(s)
- Charlotte Taylor
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - David Mannion
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Fabrizio Miranda
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Mohammad Karaminejadranjbar
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Sandra Herrero-Gonzalez
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Karin Hellner
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Yiyan Zheng
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Geoffrey Bartholomeusz
- Department of Experimental Therapeutics, M.D. Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Robert C Bast
- Department of Experimental Therapeutics, M.D. Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Ahmed Ashour Ahmed
- Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, OX3 9DS, UK.,Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| |
Collapse
|
28
|
Ciarlo C, Kaufman CK, Kinikoglu B, Michael J, Yang S, D′Amato C, Blokzijl-Franke S, den Hertog J, Schlaeger TM, Zhou Y, Liao E, Zon LI. A chemical screen in zebrafish embryonic cells establishes that Akt activation is required for neural crest development. eLife 2017; 6:e29145. [PMID: 28832322 PMCID: PMC5599238 DOI: 10.7554/elife.29145] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/08/2017] [Indexed: 01/09/2023] Open
Abstract
The neural crest is a dynamic progenitor cell population that arises at the border of neural and non-neural ectoderm. The inductive roles of FGF, Wnt, and BMP at the neural plate border are well established, but the signals required for subsequent neural crest development remain poorly characterized. Here, we conducted a screen in primary zebrafish embryo cultures for chemicals that disrupt neural crest development, as read out by crestin:EGFP expression. We found that the natural product caffeic acid phenethyl ester (CAPE) disrupts neural crest gene expression, migration, and melanocytic differentiation by reducing Sox10 activity. CAPE inhibits FGF-stimulated PI3K/Akt signaling, and neural crest defects in CAPE-treated embryos are suppressed by constitutively active Akt1. Inhibition of Akt activity by constitutively active PTEN similarly decreases crestin expression and Sox10 activity. Our study has identified Akt as a novel intracellular pathway required for neural crest differentiation.
Collapse
Affiliation(s)
- Christie Ciarlo
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
- Harvard Medical SchoolBostonUnited States
| | - Charles K Kaufman
- Division of Oncology, Department of MedicineWashington University School of MedicineSt. LouisUnited States
- Department of Developmental BiologyWashington University School of MedicineSt. LouisUnited States
| | - Beste Kinikoglu
- Center for Regenerative MedicineMassachusetts General HospitalBostonUnited States
- Division of Plastic and Reconstructive SurgeryMassachusetts General HospitalBostonUnited States
| | - Jonathan Michael
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Song Yang
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Christopher D′Amato
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Sasja Blokzijl-Franke
- Hubrecht Institute, Koninklijke Nederlandse Akademie van WetenschappenUniversity Medical Center UtrechtUtrechtNetherlands
| | - Jeroen den Hertog
- Hubrecht Institute, Koninklijke Nederlandse Akademie van WetenschappenUniversity Medical Center UtrechtUtrechtNetherlands
| | - Thorsten M Schlaeger
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Yi Zhou
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
| | - Eric Liao
- Harvard Medical SchoolBostonUnited States
- Center for Regenerative MedicineMassachusetts General HospitalBostonUnited States
- Division of Plastic and Reconstructive SurgeryMassachusetts General HospitalBostonUnited States
- Harvard Stem Cell InstituteCambridgeUnited States
| | - Leonard I Zon
- Stem Cell Program and Hematology/OncologyChildren’s Hospital Boston, Howard Hughes Medical InstituteBostonUnited States
- Harvard Medical SchoolBostonUnited States
- Harvard Stem Cell InstituteCambridgeUnited States
| |
Collapse
|
29
|
Figueiredo AL, Maczkowiak F, Borday C, Pla P, Sittewelle M, Pegoraro C, Monsoro-Burq AH. PFKFB4 control of Akt signaling is essential for premigratory and migratory neural crest formation. Development 2017; 144:4183-4194. [DOI: 10.1242/dev.157644] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/07/2017] [Indexed: 01/05/2023]
Abstract
Neural crest (NC) specification comprises an early phase, initiating immature NC progenitors formation at neural plate stage, and a later phase at neural fold stage, resulting into functional premigratory NC, able to delaminate and migrate. We found that the NC Gene Regulatory Network triggers up-regulation of pfkfb4 (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 4) during this late specification phase. As shown in previous studies, PFKFB4 controls AKT signaling in gastrulas and glycolysis rate in adult cells. Here, we focus on PFKFB4 function in NC during and after neurulation, using time-controlled or hypomorph depletions in vivo. We find that PFKFB4 is essential both for specification of functional premigratory NC and for its migration. PFKFB4-depleted embryos fail activating n-cadherin and late NC specifiers, exhibit severe migration defects, resulting in craniofacial defects. AKT signaling mediates PFKFB4 function in NC late specification, while both AKT signaling and glycolysis regulate migration. These findings highlight novel and critical roles of PFKFB4 activity in later stages of NC development, wired into the NC-GRN.
Collapse
Affiliation(s)
- Ana Leonor Figueiredo
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405, Orsay, France
- Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, France
| | - Frédérique Maczkowiak
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405, Orsay, France
- Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, France
| | - Caroline Borday
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405, Orsay, France
- Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, France
| | - Patrick Pla
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405, Orsay, France
- Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, France
| | - Meghane Sittewelle
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405, Orsay, France
- Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, France
| | - Caterina Pegoraro
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405, Orsay, France
- Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, France
| | - Anne H. Monsoro-Burq
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405, Orsay, France
- Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, France
- Institut Universitaire de France, Paris, France
| |
Collapse
|