101
|
Systemic Activation of Activin A Signaling Causes Chronic Kidney Disease-Mineral Bone Disorder. Int J Mol Sci 2018; 19:ijms19092490. [PMID: 30142896 PMCID: PMC6163495 DOI: 10.3390/ijms19092490] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 11/19/2022] Open
Abstract
The high cardiovascular mortality associated with chronic kidney disease (CKD) is caused in part by the CKD-mineral bone disorder (CKD-MBD) syndrome. The CKD-MBD consists of skeletal, vascular and cardiac pathology caused by metabolic derangements produced by kidney disease. The prevalence of osteopenia/osteoporosis resulting from the skeletal component of the CKD-MBD, renal osteodystrophy (ROD), in patients with CKD exceeds that of the general population and is a major public health concern. That CKD is associated with compromised bone health is widely accepted, yet the mechanisms underlying impaired bone metabolism in CKD are not fully understood. Therefore, clarification of the molecular mechanisms by which CKD produces ROD is of crucial significance. We have shown that activin A, a member of the transforming growth factor (TGF)-β super family, is an important positive regulator of receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclastogenesis with Smad-mediated signaling being crucial for inducing osteoclast development and function. Recently, we have demonstrated systemic activation of activin receptors and activin A levels in CKD mouse models, such as diabetic CKD and Alport (AL) syndrome. In these CKD mouse models, bone remodeling caused by increased osteoclast numbers and activated osteoclastic bone resorption was observed and treatment with an activin receptor ligand trap repaired CKD-induced-osteoclastic bone resorption and stimulated individual osteoblastic bone formation, irrespective of parathyroid hormone (PTH) elevation. These findings have opened a new field for exploring mechanisms of activin A-enhanced osteoclast formation and function in CKD. Activin A appears to be a strong candidate for CKD-induced high-turnover ROD. Therefore, the treatment with the decoy receptor for activin A might be a good candidate for treatment for CKD-induced osteopenia or osteoporosis, indicating that the new findings from in these studies will lead to the identification of novel therapeutic targets for CKD-related and osteopenia and osteoporosis in general. In this review, we describe the impact of CKD-induced Smad signaling in osteoclasts, osteoblasts and vascular cells in CKD.
Collapse
|
102
|
Bragança J. SMAD2/3, versatile molecular tools for cellular engineering. Stem Cell Investig 2018; 5:24. [PMID: 30148157 PMCID: PMC6088207 DOI: 10.21037/sci.2018.07.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 07/17/2018] [Indexed: 01/01/2023]
Affiliation(s)
- José Bragança
- Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal
- Centre for Biomedical Research-CBMR, University of Algarve, Campus of Gambelas, 8005-139 Faro, Portugal
- ABC - Algarve Biomedical Centre, 8005-139 Faro, Portugal
| |
Collapse
|
103
|
David CJ, Massagué J. Contextual determinants of TGFβ action in development, immunity and cancer. Nat Rev Mol Cell Biol 2018; 19:419-435. [PMID: 29643418 DOI: 10.1038/s41580-018-0007-0] [Citation(s) in RCA: 596] [Impact Index Per Article: 85.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Few cell signals match the impact of the transforming growth factor-β (TGFβ) family in metazoan biology. TGFβ cytokines regulate cell fate decisions during development, tissue homeostasis and regeneration, and are major players in tumorigenesis, fibrotic disorders, immune malfunctions and various congenital diseases. The effects of the TGFβ family are mediated by a combinatorial set of ligands and receptors and by a common set of receptor-activated mothers against decapentaplegic homologue (SMAD) transcription factors, yet the effects can differ dramatically depending on the cell type and the conditions. Recent progress has illuminated a model of TGFβ action in which SMADs bind genome-wide in partnership with lineage-determining transcription factors and additionally integrate inputs from other pathways and the chromatin to trigger specific cellular responses. These new insights clarify the operating logic of the TGFβ pathway in physiology and disease.
Collapse
Affiliation(s)
- Charles J David
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Tsinghua University School of Medicine, Department of Basic Sciences, Beijing, China
| | - Joan Massagué
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
104
|
Yakhkeshi S, Rahimi S, Sharafi M, Hassani S, Taleahmad S, Shahverdi A, Baharvand H. In vitro improvement of quail primordial germ cell expansion through activation of TGF‐beta signaling pathway. J Cell Biochem 2018; 119:4309-4319. [DOI: 10.1002/jcb.26618] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/12/2017] [Indexed: 01/17/2023]
Affiliation(s)
- Saeed Yakhkeshi
- Department of Poultry ScienceFaculty of AgricultureTarbiat Modares UniversityTehranIran
| | - Shaban Rahimi
- Department of Poultry ScienceFaculty of AgricultureTarbiat Modares UniversityTehranIran
| | - Mohsen Sharafi
- Department of Poultry ScienceFaculty of AgricultureTarbiat Modares UniversityTehranIran
| | - Seyedeh‐Nafiseh Hassani
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Sara Taleahmad
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Abdolhossein Shahverdi
- Department of EmbryologyReproductive Biomedicine Research CenterRoyan Institute for Reproductive Biomedicine, ACECRTehranIran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental BiologyCell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Department of Developmental BiologyUniversity of Science and CultureTehranIran
| |
Collapse
|
105
|
Fernández-Pérez D, Brieño-Enríquez MA, Isoler-Alcaraz J, Larriba E, Del Mazo J. MicroRNA dynamics at the onset of primordial germ and somatic cell sex differentiation during mouse embryonic gonad development. RNA (NEW YORK, N.Y.) 2018; 24:287-303. [PMID: 29187591 PMCID: PMC5824349 DOI: 10.1261/rna.062869.117] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 11/27/2017] [Indexed: 06/07/2023]
Abstract
In mammals, commitment and specification of germ cell lines involves complex programs that include sex differentiation, control of proliferation, and meiotic initiation. Regulation of these processes is genetically controlled by fine-tuned mechanisms of gene regulation in which microRNAs (miRNAs) are involved. We have characterized, by small-RNA-seq and bioinformatics analyses, the miRNA expression patterns of male and female mouse primordial germ cells (PGCs) and gonadal somatic cells at embryonic stages E11.5, E12.5, and E13.5. Differential expression analyses revealed differences in the regulation of key miRNA clusters such as miR-199-214, miR-182-183-96, and miR-34c-5p, whose targets have defined roles during gonadal sexual determination in both germ and somatic cells. Extensive analyses of miRNA sequences revealed an increase in noncanonical isoforms on PGCs at E12.5 and dramatic changes of 3' isomiR expression and 3' nontemplate nucleotide additions in female PGCs at E13.5. Additionally, RT-qPCR analyses of genes encoding proteins involved in miRNA biogenesis and 3' nucleotide addition uncovered sexually and developmentally specific expression, characterized by the decay of Drosha, Dgcr8, and Xpo5 expression along gonadal development. These results demonstrate that miRNAs, their isomiRs, and miRNA machinery are differentially regulated and participate actively in gonadal sexual differentiation in both PGCs and gonadal somatic cells.
Collapse
Affiliation(s)
- Daniel Fernández-Pérez
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CSIC), Madrid 28040, Spain
| | - Miguel A Brieño-Enríquez
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CSIC), Madrid 28040, Spain
| | - Javier Isoler-Alcaraz
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CSIC), Madrid 28040, Spain
| | - Eduardo Larriba
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CSIC), Madrid 28040, Spain
| | - Jesús Del Mazo
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CSIC), Madrid 28040, Spain
| |
Collapse
|
106
|
Triggering the activation of Activin A type II receptor in human adipose stem cells towards tenogenic commitment using mechanomagnetic stimulation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1149-1159. [PMID: 29471171 DOI: 10.1016/j.nano.2018.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/16/2018] [Accepted: 02/10/2018] [Indexed: 12/21/2022]
Abstract
Stem cell therapies hold potential to stimulate tendon regeneration and homeostasis, which is maintained in response to the native mechanical environment. Activins are members of the mechano-responsive TGF-β superfamily that participates in the regulation of several downstream biological processes. Mechanosensitive membrane receptors such as activin can be activated in different types of stem cells via magnetic nanoparticles (MNPs) through remote magnetic actuation resulting in cell differentiation. In this work, we target the Activin receptor type IIA (ActRIIA) in human adipose stem cells (hASCs), using anti-ActRIIA functionalized MNPs, externally activated through a oscillating magnetic bioreactor. Upon activation, the phosphorylation of Smad2/3 is induced allowing translocation of the complex to the nucleus, regulating tenogenic transcriptional responses. Our study demonstrates the potential remote activation of MNPs tagged hASCs to trigger the Activin receptor leading to tenogenic differentiation. These results may provide insights toward tendon regeneration therapies.
Collapse
|
107
|
Laminin 521 Stabilizes the Pluripotency Expression Pattern of Human Embryonic Stem Cells Initially Derived on Feeder Cells. Stem Cells Int 2018. [PMID: 29535778 PMCID: PMC5835285 DOI: 10.1155/2018/7127042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Human embryonic stem (hES) cells represent an important tool to study early cell development. The previously described use of human recombinant laminin (LN) 521 represented a step forward in generating clinically safe culture conditions. To test the short-term effect of LN521 on cultured hES cells, five male hES cell lines were cultured on human foreskin fibroblasts (hFFs), Matrigel, LN521, and LN121 and characterized by qPCR, immunofluorescence analysis, as well as their potential for three-germ layer differentiation. Variations in gene expression related to pluripotency, stemness, and testicular cells at different passages and culture conditions were evaluated by qPCR. All cell lines expressed pluripotency markers at protein and RNA level and were able to differentiate into cell types of the three germ layers after being cultured on LN521 for nine passages. Reduction in variation of pluripotency marker expression could be observed after culturing the cells on LN521 for nine passages. hES cells cultured on LN521 exhibited less differentiation, faster cell growth, and attachment when compared to hES cells cultured on LN121 or Matrigel. Our results indicate a positive effect of LN521 in stabilizing pluripotency gene expression and might be the first step towards more controllable and robust culture conditions for hES cells.
Collapse
|
108
|
Signore IA, Palma K, Concha ML. Nodal signalling and asymmetry of the nervous system. Philos Trans R Soc Lond B Biol Sci 2017; 371:rstb.2015.0401. [PMID: 27821531 DOI: 10.1098/rstb.2015.0401] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2016] [Indexed: 11/12/2022] Open
Abstract
The role of Nodal signalling in nervous system asymmetry is still poorly understood. Here, we review and discuss how asymmetric Nodal signalling controls the ontogeny of nervous system asymmetry using a comparative developmental perspective. A detailed analysis of asymmetry in ascidians and fishes reveals a critical context-dependency of Nodal function and emphasizes that bilaterally paired and midline-unpaired structures/organs behave as different entities. We propose a conceptual framework to dissect the developmental function of Nodal as asymmetry inducer and laterality modulator in the nervous system, which can be used to study other types of body and visceral organ asymmetries. Using insights from developmental biology, we also present novel evolutionary hypotheses on how Nodal led the evolution of directional asymmetry in the brain, with a particular focus on the epithalamus. We intend this paper to provide a synthesis on how Nodal signalling controls left-right asymmetry of the nervous system.This article is part of the themed issue 'Provocative questions in left-right asymmetry'.
Collapse
Affiliation(s)
- Iskra A Signore
- Anatomy and Developmental Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, PO Box 70031, Santiago, Chile.,Biomedical Neuroscience Institute, Independencia 1027, Santiago, Chile
| | - Karina Palma
- Anatomy and Developmental Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, PO Box 70031, Santiago, Chile.,Biomedical Neuroscience Institute, Independencia 1027, Santiago, Chile
| | - Miguel L Concha
- Anatomy and Developmental Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, PO Box 70031, Santiago, Chile .,Biomedical Neuroscience Institute, Independencia 1027, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| |
Collapse
|
109
|
Collinson A, Collier AJ, Morgan NP, Sienerth AR, Chandra T, Andrews S, Rugg-Gunn PJ. Deletion of the Polycomb-Group Protein EZH2 Leads to Compromised Self-Renewal and Differentiation Defects in Human Embryonic Stem Cells. Cell Rep 2017; 17:2700-2714. [PMID: 27926872 PMCID: PMC5177603 DOI: 10.1016/j.celrep.2016.11.032] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 10/11/2016] [Accepted: 11/09/2016] [Indexed: 12/11/2022] Open
Abstract
Through the histone methyltransferase EZH2, the Polycomb complex PRC2 mediates H3K27me3 and is associated with transcriptional repression. PRC2 regulates cell-fate decisions in model organisms; however, its role in regulating cell differentiation during human embryogenesis is unknown. Here, we report the characterization of EZH2-deficient human embryonic stem cells (hESCs). H3K27me3 was lost upon EZH2 deletion, identifying an essential requirement for EZH2 in methylating H3K27 in hESCs, in contrast to its non-essential role in mouse ESCs. Developmental regulators were derepressed in EZH2-deficient hESCs, and single-cell analysis revealed an unexpected acquisition of lineage-restricted transcriptional programs. EZH2-deficient hESCs show strongly reduced self-renewal and proliferation, thereby identifying a more severe phenotype compared to mouse ESCs. EZH2-deficient hESCs can initiate differentiation toward developmental lineages; however, they cannot fully differentiate into mature specialized tissues. Thus, EZH2 is required for stable ESC self-renewal, regulation of transcriptional programs, and for late-stage differentiation in this model of early human development. Comprehensive examination of EZH2 function in human ESC regulation EZH2 deficiency causes lineage-restricted derepression of developmental regulators More severe self-renewal and growth defects in EZH2-deficient hESCs than in mESCs EZH2-deficient hESCs can differentiate to early lineages but cannot form mature tissues
Collapse
Affiliation(s)
- Adam Collinson
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Amanda J Collier
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK; Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK
| | - Natasha P Morgan
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Arnold R Sienerth
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Tamir Chandra
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Simon Andrews
- Bioinformatics Group, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Peter J Rugg-Gunn
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK; Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
110
|
Montague TG, Schier AF. Vg1-Nodal heterodimers are the endogenous inducers of mesendoderm. eLife 2017; 6:28183. [PMID: 29140251 PMCID: PMC5745085 DOI: 10.7554/elife.28183] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 10/13/2017] [Indexed: 12/03/2022] Open
Abstract
Nodal is considered the key inducer of mesendoderm in vertebrate embryos and embryonic stem cells. Other TGF-beta-related signals, such as Vg1/Dvr1/Gdf3, have also been implicated in this process but their roles have been unclear or controversial. Here we report that zebrafish embryos without maternally provided vg1 fail to form endoderm and head and trunk mesoderm, and closely resemble nodal loss-of-function mutants. Although Nodal is processed and secreted without Vg1, it requires Vg1 for its endogenous activity. Conversely, Vg1 is unprocessed and resides in the endoplasmic reticulum without Nodal, and is only secreted, processed and active in the presence of Nodal. Co-expression of Nodal and Vg1 results in heterodimer formation and mesendoderm induction. Thus, mesendoderm induction relies on the combination of two TGF-beta-related signals: maternal and ubiquitous Vg1, and zygotic and localized Nodal. Modeling reveals that the pool of maternal Vg1 enables rapid signaling at low concentrations of zygotic Nodal. All animals begin life as just one cell – a fertilized egg. In order to make a recognizable adult, each embryo needs to make the three types of tissue that will eventually form all of the organs: endoderm, which will form the internal organs; mesoderm, which will form the muscle and bones; and ectoderm, which will generate the skin and nervous system. All vertebrates – animals with backbones like fish and humans – use the so-called Nodal signaling pathway to make the endoderm and mesoderm. Nodal is a signaling molecule that binds to receptors on the surface of cells. If Nodal binds to a receptor on a cell, it instructs that cell to become endoderm or mesoderm. As such, Nodal is critical for vertebrate life. However, there has been a 30-year debate in the field of developmental biology about whether a protein called Vg1, which has a similar molecular structure as Nodal, plays a role in the early development of vertebrates. Zebrafish are often used to study animal development, and Montague and Schier decided to test whether these fish need the gene for Vg1 (also known as Gdf3) by deleting it using a genome editing technique called CRISPR/Cas9. It turns out that female zebrafish can survive without this gene. Yet, when the offspring of these females do not inherit the instructions to make Vg1 from their mothers, they fail to form the endoderm and mesoderm. This means that the embryos do not have hearts, blood or other internal organs, and they die within three days. Two other groups of researchers have independently reported similar results. The findings reveal that Vg1 is critical for the Nodal signaling pathway to work in zebrafish. Montague and Schier then showed that, in this pathway, Nodal does not activate its receptors on its own. Instead, Nodal must interact with Vg1, and it is this Nodal-Vg1 complex that activates receptors, and instructs cells to become endoderm and mesoderm. Scientists currently use the Nodal signaling pathway to induce human embryonic stem cells growing in the laboratory to become mesoderm and endoderm. As such, these new findings could ultimately help researchers to grow tissues and organs for human patients.
Collapse
Affiliation(s)
- Tessa G Montague
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Alexander F Schier
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States.,Center for Brain Science, Harvard University, Cambridge, United States.,Broad Institute of MIT and Harvard, Cambridge, United States.,Harvard Stem Cell Institute, Cambridge, United States.,FAS Center for Systems Biology, Harvard University, Cambridge, United States
| |
Collapse
|
111
|
MacFarlane EG, Haupt J, Dietz HC, Shore EM. TGF-β Family Signaling in Connective Tissue and Skeletal Diseases. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022269. [PMID: 28246187 DOI: 10.1101/cshperspect.a022269] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The transforming growth factor β (TGF-β) family of signaling molecules, which includes TGF-βs, activins, inhibins, and numerous bone morphogenetic proteins (BMPs) and growth and differentiation factors (GDFs), has important functions in all cells and tissues, including soft connective tissues and the skeleton. Specific TGF-β family members play different roles in these tissues, and their activities are often balanced with those of other TGF-β family members and by interactions with other signaling pathways. Perturbations in TGF-β family pathways are associated with numerous human diseases with prominent involvement of the skeletal and cardiovascular systems. This review focuses on the role of this family of signaling molecules in the pathologies of connective tissues that manifest in rare genetic syndromes (e.g., syndromic presentations of thoracic aortic aneurysm), as well as in more common disorders (e.g., osteoarthritis and osteoporosis). Many of these diseases are caused by or result in pathological alterations of the complex relationship between the TGF-β family of signaling mediators and the extracellular matrix in connective tissues.
Collapse
Affiliation(s)
- Elena Gallo MacFarlane
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Julia Haupt
- Department of Orthopedic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Center for Research in FOP and Related Disorders, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Harry C Dietz
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205.,Howard Hughes Medical Institute, Bethesda, Maryland 21205
| | - Eileen M Shore
- Department of Orthopedic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Center for Research in FOP and Related Disorders, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
112
|
Calvanese L, Focà A, Sandomenico A, Focà G, Caporale A, Doti N, Iaccarino E, Leonardi A, D'Auria G, Ruvo M, Falcigno L. Structural insights into the interaction of a monoclonal antibody and Nodal peptides by STD-NMR spectroscopy. Bioorg Med Chem 2017; 25:6589-6596. [PMID: 29113739 DOI: 10.1016/j.bmc.2017.10.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/05/2017] [Accepted: 10/26/2017] [Indexed: 12/31/2022]
Abstract
Nodal is a growth factor expressed during early embryonic development, but reactivated in several advanced-stage cancers. Targeting of Nodal signaling, which occurs via the binding to Cripto-1 co-receptor, results in inhibition of cell aggressiveness and reduced tumor growth. The Nodal binding region to Cripto-1 was identified and targeted with a high affinity monoclonal antibody (3D1). By STD-NMR technique, we investigated the interaction of Nodal fragments with 3D1 with the aim to elucidate at atomic level the interaction surface. Data indicate with high accuracy the antibody-antigen contact atoms and confirm the information previously obtained by immune-enzymatic methods. Main residues contacted by 3D1 are P46, V47, E49 and E50, which belong to the Nodal loop involved in the interaction with the co-receptor.
Collapse
Affiliation(s)
- Luisa Calvanese
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Annalia Focà
- Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Annamaria Sandomenico
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy; Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Giuseppina Focà
- Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Andrea Caporale
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Nunzianna Doti
- Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Emanuela Iaccarino
- DISTABIF, Università degli Studi della Campania "Lugi Vanvitelli", via Vivaldi, 43, 80100 Caserta, Italy
| | - Antonio Leonardi
- Dept. Medicina Molecolare e Biotecnologie Mediche, Università Federico II di Napoli, Naples, Italy
| | - Gabriella D'Auria
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy; Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy; Dept. of Pharmacy, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Menotti Ruvo
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy; Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy.
| | - Lucia Falcigno
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy; Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy; Dept. of Pharmacy, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy.
| |
Collapse
|
113
|
Xiang J, Cao S, Zhong L, Wang H, Pei Y, Wei Q, Wen B, Mu H, Zhang S, Yue L, Yue G, Lim B, Han J. Pluripotent stem cells secrete Activin A to improve their epiblast competency after injection into recipient embryos. Protein Cell 2017; 9:717-728. [PMID: 29027123 PMCID: PMC6053354 DOI: 10.1007/s13238-017-0470-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/22/2017] [Indexed: 11/10/2022] Open
Abstract
It is not fully clear why there is a higher contribution of pluripotent stem cells (PSCs) to the chimera produced by injection of PSCs into 4-cell or 8-cell stage embryos compared with blastocyst injection. Here, we show that not only embryonic stem cells (ESCs) but also induced pluripotent stem cells (iPSCs) can generate F0 nearly 100% donor cell-derived mice by 4-cell stage embryo injection, and the approach has a “dose effect”. Through an analysis of the PSC-secreted proteins, Activin A was found to impede epiblast (EPI) lineage development while promoting trophectoderm (TE) differentiation, resulting in replacement of the EPI lineage of host embryos with PSCs. Interestingly, the injection of ESCs into blastocysts cultured with Activin A (cultured from 4-cell stage to early blastocyst at E3.5) could increase the contribution of ESCs to the chimera. The results indicated that PSCs secrete protein Activin A to improve their EPI competency after injection into recipient embryos through influencing the development of mouse early embryos. This result is useful for optimizing the chimera production system and for a deep understanding of PSCs effects on early embryo development.
Collapse
Affiliation(s)
- Jinzhu Xiang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Suying Cao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Liang Zhong
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Hanning Wang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China
| | - Yangli Pei
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China.,State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Qingqing Wei
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Bingqiang Wen
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Haiyuan Mu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Shaopeng Zhang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Liang Yue
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Genhua Yue
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, 117604, Singapore
| | - Bing Lim
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Jianyong Han
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
114
|
Jiang L, Si T, Yu M, Zeng X, Morse HC, Lu Y, Ouyang G, Zhou JX. The tumor suppressive role of inhibin βA in diffuse large B-cell lymphoma. Leuk Lymphoma 2017; 59:1202-1212. [PMID: 28877610 DOI: 10.1080/10428194.2017.1372574] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INHBA (inhibin βA), a subunit of a ligand of the transforming growth factor-β superfamily, is known to play diverse roles in various solid tumors. However, its role in hematologic malignancies remains unexplored. Here, we investigated the function of INHBA in diffuse large B-cell lymphoma (DLBCL). Both mRNA and protein levels of INHBA were significantly downregulated in primary DLBCL tissues, irrespective of germinal center B-cell-like (GCB) or non-GCB subtype, compared to those in benign tonsils. The low level of INHBA in patients with de novo DLBCL was correlated with reduced overall and progression-free survival. Ectopic expression of INHBA in DLBCL cell lines (OCI-Ly01 and SUDHL-10) resulted in reduced cell proliferation, increased spontaneous apoptosis and arrested cell cycle in vitro and suppressed xenograft tumor growth in vivo. Moreover, INHBA enhanced the chemosensitivity of DLBCL cells. Thus, our results provide novel evidence that INHBA functions as a tumor suppressor in DLBCL.
Collapse
Affiliation(s)
- Lei Jiang
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China
| | - Ting Si
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China.,b Department of Hematology , Affiliated Yinzhou Hospital, Ningbo University School of Medicine , Ningbo , China
| | - Mei Yu
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China
| | - Xinli Zeng
- c Department of ENT , Ningbo Second People's Hospital , Ningbo , China
| | - Herbert C Morse
- d Laboratory of Immunopathology , National Institute of Allergy and Infectious Diseases, National Institutes of Health , Rockville , MD , USA
| | - Ying Lu
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China.,b Department of Hematology , Affiliated Yinzhou Hospital, Ningbo University School of Medicine , Ningbo , China
| | - Guifang Ouyang
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China.,e Department of Hematology , Ningbo First People's Hospital , Ningbo , China
| | - Jeff X Zhou
- a Department of Pathology, Zhejiang Provincial Key Laboratory of Pathophysiology , Ningbo University School of Medicine , Ningbo , China
| |
Collapse
|
115
|
Margaryan NV, Seftor EA, Seftor RE, Hendrix MJ. Targeting the Stem Cell Properties of Adult Breast Cancer Cells: Using Combinatorial Strategies to Overcome Drug Resistance. CURRENT MOLECULAR BIOLOGY REPORTS 2017; 3:159-164. [PMID: 29152453 PMCID: PMC5687579 DOI: 10.1007/s40610-017-0067-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW Cancer is a major public health problem worldwide. In aggressive cancers, which are heterogeneous in nature, there exists a paucity of targetable molecules that can be used to predict outcome and response to therapy in patients, especially those in the high risk category with a propensity to relapse following chemotherapy. This review addresses the challenges pertinent to treating aggressive cancer cells with inherent stem cell properties, with a special focus on triple-negative breast cancer (TNBC). RECENT FINDINGS Plasticity underlies the cancer stem cell (CSC) phenotype in aggressive cancers like TNBC. Progenitors and CSCs implement similar signaling pathways to sustain growth, and the convergence of embryonic and tumorigenic signaling pathways has led to the discovery of novel oncofetal targets, rigorously regulated during normal development, but aberrantly reactivated in aggressive forms of cancer. SUMMARY Translational studies have shown that Nodal, an embryonic morphogen, is reactivated in aggressive cancers, but not in normal tissues, and underlies tumor growth, invasion, metastasis and drug resistance. Front-line therapies do not inhibit Nodal, but when a combinatorial approach is used with an agent such as doxorubicin followed by anti-Nodal antibody therapy, significant decreases in cell growth and viability occur. These findings are of special interest in the development of new therapeutic interventions that target the stem cell properties of cancer cells to overcome drug resistance and metastasis.
Collapse
Affiliation(s)
- Naira V. Margaryan
- Department of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506 USA
- Cancer Institute, West Virginia University, Morgantown, WV 26506 USA
- Department of Biology, Shepherd University, Shepherdstown, WV 25443 USA
| | - Elisabeth A. Seftor
- Department of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506 USA
- Cancer Institute, West Virginia University, Morgantown, WV 26506 USA
- Department of Biology, Shepherd University, Shepherdstown, WV 25443 USA
| | - Richard E.B. Seftor
- Department of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506 USA
- Cancer Institute, West Virginia University, Morgantown, WV 26506 USA
- Department of Biology, Shepherd University, Shepherdstown, WV 25443 USA
| | - Mary J.C. Hendrix
- Department of Internal Medicine, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506 USA
- Cancer Institute, West Virginia University, Morgantown, WV 26506 USA
- Department of Biology, Shepherd University, Shepherdstown, WV 25443 USA
| |
Collapse
|
116
|
Lu AQ, Popova EY, Barnstable CJ. Activin Signals through SMAD2/3 to Increase Photoreceptor Precursor Yield during Embryonic Stem Cell Differentiation. Stem Cell Reports 2017; 9:838-852. [PMID: 28781074 PMCID: PMC5599185 DOI: 10.1016/j.stemcr.2017.06.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 01/23/2023] Open
Abstract
In vitro differentiation of mouse embryonic stem cells (ESCs) into retinal fates can be used to study the roles of exogenous factors acting through multiple signaling pathways during retina development. Application of activin A during a specific time frame that corresponds to early embryonic retinogenesis caused increased generation of CRX+ photoreceptor precursors and decreased PAX6+ retinal progenitor cells (RPCs). Following activin A treatment, SMAD2/3 was activated in RPCs and bound to promoter regions of key RPC and photoreceptor genes. The effect of activin on CRX expression was repressed by pharmacological inhibition of SMAD2/3 phosphorylation. Activin signaling through SMAD2/3 in RPCs regulates expression of transcription factors involved in cell type determination and promotes photoreceptor lineage specification. Our findings can contribute to the production of photoreceptors for cell replacement therapy.
Collapse
Affiliation(s)
- Amy Q Lu
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Evgenya Y Popova
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Colin J Barnstable
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
117
|
Future Challenges in the Generation of Hepatocyte-Like Cells From Human Pluripotent Stem Cells. CURRENT PATHOBIOLOGY REPORTS 2017. [DOI: 10.1007/s40139-017-0150-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
118
|
Abstract
Recent breakthroughs in pluripotent stem cell technologies have enabled a new class of in vitro systems for functional modeling of human brain development. These advances, in combination with improvements in neural differentiation methods, allow the generation of in vitro systems that reproduce many in vivo features of the brain with remarkable similarity. Here, we describe advances in the development of these methods, focusing on neural rosette and organoid approaches, and compare their relative capabilities and limitations. We also discuss current technical hurdles for recreating the cell-type complexity and spatial architecture of the brain in culture and offer potential solutions.
Collapse
|
119
|
Kloesel B, DiNardo JA, Body SC. Cardiac Embryology and Molecular Mechanisms of Congenital Heart Disease: A Primer for Anesthesiologists. Anesth Analg 2017; 123:551-69. [PMID: 27541719 DOI: 10.1213/ane.0000000000001451] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Congenital heart disease is diagnosed in 0.4% to 5% of live births and presents unique challenges to the pediatric anesthesiologist. Furthermore, advances in surgical management have led to improved survival of those patients, and many adult anesthesiologists now frequently take care of adolescents and adults who have previously undergone surgery to correct or palliate congenital heart lesions. Knowledge of abnormal heart development on the molecular and genetic level extends and improves the anesthesiologist's understanding of congenital heart disease. In this article, we aim to review current knowledge pertaining to genetic alterations and their cellular effects that are involved in the formation of congenital heart defects. Given that congenital heart disease can currently only occasionally be traced to a single genetic mutation, we highlight some of the difficulties that researchers face when trying to identify specific steps in the pathogenetic development of heart lesions.
Collapse
Affiliation(s)
- Benjamin Kloesel
- From the Department of Anesthesia, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | | | | |
Collapse
|
120
|
Bodenstine TM, Chandler GS, Reed DW, Margaryan NV, Gilgur A, Atkinson J, Ahmed N, Hyser M, Seftor EA, Strizzi L, Hendrix MJC. Nodal expression in triple-negative breast cancer: Cellular effects of its inhibition following doxorubicin treatment. Cell Cycle 2017; 15:1295-302. [PMID: 27007464 DOI: 10.1080/15384101.2016.1160981] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Triple-negative breast cancer (TNBC) represents an aggressive cancer subtype characterized by the lack of expression of estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2). The independence of TNBC from these growth promoting factors eliminates the efficacy of therapies which specifically target them, and limits TNBC patients to traditional systemic neo/adjuvant chemotherapy. To better understand the growth advantage of TNBC - in the absence of ER, PR and HER2, we focused on the embryonic morphogen Nodal (associated with the cancer stem cell phenotype), which is re-expressed in aggressive breast cancers. Most notably, our previous data demonstrated that inhibition of Nodal signaling in breast cancer cells reduces their tumorigenic capacity. Furthermore, inhibiting Nodal in other cancers has resulted in improved effects of chemotherapy, although the mechanisms for this remain unknown. Thus, we hypothesized that targeting Nodal in TNBC cells in combination with conventional chemotherapy may improve efficacy and represent a potential new strategy. Our preliminary data demonstrate that Nodal is highly expressed in TNBC when compared to invasive hormone receptor positive samples. Treatment of Nodal expressing TNBC cell lines with a neutralizing anti-Nodal antibody reduces the viability of cells that had previously survived treatment with the anthracycline doxorubicin. We show that inhibiting Nodal may alter response mechanisms employed by cancer cells undergoing DNA damage. These data suggest that development of therapies which target Nodal in TNBC may lead to additional treatment options in conjunction with chemotherapy regimens - by altering signaling pathways critical to cellular survival.
Collapse
Affiliation(s)
- Thomas M Bodenstine
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA
| | - Grace S Chandler
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA
| | - David W Reed
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA
| | - Naira V Margaryan
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA
| | - Alina Gilgur
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA
| | | | - Nida Ahmed
- b Presence Saint Francis Hospital , Evanston , IL , USA
| | - Matthew Hyser
- b Presence Saint Francis Hospital , Evanston , IL , USA
| | - Elisabeth A Seftor
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA
| | - Luigi Strizzi
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA.,c Department of Pathology , Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| | - Mary J C Hendrix
- a Cancer Biology and Epigenomics Program at the Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago , Chicago , IL , USA.,d Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| |
Collapse
|
121
|
Farnsworth DR, Doe CQ. Opportunities lost and gained: Changes in progenitor competence during nervous system development. NEUROGENESIS 2017; 4:e1324260. [PMID: 28656157 DOI: 10.1080/23262133.2017.1324260] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 04/20/2017] [Accepted: 04/24/2017] [Indexed: 02/07/2023]
Abstract
During development of the central nervous system, a small pool of stem cells and progenitors generate the vast neural diversity required for neural circuit formation and behavior. Neural stem and progenitor cells often generate different progeny in response to the same signaling cue (e.g. Notch or Hedgehog), including no response at all. How does stem cell competence to respond to signaling cues change over time? Recently, epigenetics particularly chromatin remodeling - has emerged as a powerful mechanism to control stem cell competence. Here we review recent Drosophila and vertebrate literature describing the effect of epigenetic changes on neural stem cell competence.
Collapse
Affiliation(s)
- Dylan R Farnsworth
- Howard Hughes Medical Institute, University of Oregon, Eugene, OR, USA.,Institute of Molecular Biology, University of Oregon, Eugene, OR, USA
| | - Chris Q Doe
- Howard Hughes Medical Institute, University of Oregon, Eugene, OR, USA.,Institute of Molecular Biology, University of Oregon, Eugene, OR, USA.,Institute of Neuroscience, University of Oregon, Eugene, OR, USA
| |
Collapse
|
122
|
Song W, Cheng D, Hong S, Sappe B, Hu Y, Wei N, Zhu C, O'Connor MB, Pissios P, Perrimon N. Midgut-Derived Activin Regulates Glucagon-like Action in the Fat Body and Glycemic Control. Cell Metab 2017; 25:386-399. [PMID: 28178568 PMCID: PMC5373560 DOI: 10.1016/j.cmet.2017.01.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/03/2016] [Accepted: 01/04/2017] [Indexed: 01/08/2023]
Abstract
While high-caloric diet impairs insulin response to cause hyperglycemia, whether and how counter-regulatory hormones are modulated by high-caloric diet is largely unknown. We find that enhanced response of Drosophila adipokinetic hormone (AKH, the glucagon homolog) in the fat body is essential for hyperglycemia associated with a chronic high-sugar diet. We show that the activin type I receptor Baboon (Babo) autonomously increases AKH signaling without affecting insulin signaling in the fat body via, at least, increase of Akh receptor (AkhR) expression. Further, we demonstrate that Activin-β (Actβ), an activin ligand predominantly produced in the enteroendocrine cells (EEs) of the midgut, is upregulated by chronic high-sugar diet and signals through Babo to promote AKH action in the fat body, leading to hyperglycemia. Importantly, activin signaling in mouse primary hepatocytes also increases glucagon response and glucagon-induced glucose production, indicating a conserved role for activin in enhancing AKH/glucagon signaling and glycemic control.
Collapse
Affiliation(s)
- Wei Song
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| | - Daojun Cheng
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
| | - Shangyu Hong
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Benoit Sappe
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Yanhui Hu
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Neil Wei
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Changqi Zhu
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael B O'Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Pavlos Pissios
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| |
Collapse
|
123
|
Hendrix MJ, Kandela I, Mazar AP, Seftor EA, Seftor RE, Margaryan NV, Strizzi L, Murphy GF, Long GV, Scolyer RA. Targeting melanoma with front-line therapy does not abrogate Nodal-expressing tumor cells. J Transl Med 2017; 97:176-186. [PMID: 27775691 DOI: 10.1038/labinvest.2016.107] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/24/2016] [Accepted: 09/06/2016] [Indexed: 01/12/2023] Open
Abstract
Metastatic melanoma is a highly aggressive skin cancer with a poor prognosis. It is the leading cause of skin cancer deaths with a median overall survival for advanced-stage metastatic disease of <6 months. Despite advances in the field with conventional and targeted therapies, the heterogeneity of melanoma poses the greatest ongoing challenge, ultimately leading to relapse and progression to a more drug-resistant tumor in most patients. Particularly noteworthy are recent findings, indicating that these therapies exert selective pressure on tumors resulting in the activation of pathways associated with cancer stem cells that are unresponsive to current therapy. Our previous studies have shown how Nodal, an embryonic morphogen of the transforming growth factor-beta superfamily, is one of these critical factors that is reactivated in aggressive melanoma and resistant to conventional chemotherapy, such as dacarbazine. In the current study, we sought to determine whether BRAF inhibitor (BRAFi) therapy targeted Nodal-expressing tumor cells in uniquely matched unresectable stage III and IV melanoma patient samples before and after therapy that preceded their eventual death due to disease. The results demonstrate that BRAFi treatment failed to affect Nodal levels in melanoma tissues. Accompanying experiments in soft agar and in nude mice showed the advantage of using combinatorial treatment with BRAFi plus anti-Nodal monoclonal antibody to suppress tumor growth and metastasis. These data provide a promising new approach using front-line therapy combined with targeting a cancer stem cell-associated molecule-producing a more efficacious response than monotherapy.
Collapse
Affiliation(s)
- Mary Jc Hendrix
- Department of Biology, Shepherd University, Shepherdstown, WV, USA.,Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, USA
| | - Irawati Kandela
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
| | - Andrew P Mazar
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
| | - Elisabeth A Seftor
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, USA
| | - Richard Eb Seftor
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, USA
| | - Naira V Margaryan
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, USA
| | - Luigi Strizzi
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Pathology, Midwestern University, Downers Grove, IL, USA
| | - George F Murphy
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Georgina V Long
- Melanoma Institute Australia and Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia and Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
124
|
Luft S, Arrizabalaga O, Kulish I, Nasonova E, Durante M, Ritter S, Schroeder IS. Ionizing Radiation Alters Human Embryonic Stem Cell Properties and Differentiation Capacity by Diminishing the Expression of Activin Receptors. Stem Cells Dev 2016; 26:341-352. [PMID: 27937745 DOI: 10.1089/scd.2016.0277] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Exposure of the embryo to ionizing radiation (IR) is detrimental as it can cause genotoxic stress leading to immediate and latent consequences such as functional defects, malformations, or cancer. Human embryonic stem (hES) cells can mimic the preimplantation embryo and help to assess the biological effects of IR during early development. In this study, we describe the alterations H9 hES cells exhibit after X-ray irradiation in respect to cell cycle progression, apoptosis, genomic stability, stem cell signaling, and their capacity to differentiate into definitive endoderm. Early postirradiation, hES cells responded with an arrest in G2/M phase, elevated apoptosis, and increased chromosomal aberrations. Significant downregulation of stem cell signaling markers of the TGF beta-, Wnt-, and Hedgehog pathways was observed. Most prominent were alterations in the expression of activin receptors. However, hES cells responded differently depending on the culture conditions chosen for maintenance. Enzymatically passaged cells were less sensitive to IR than mechanically passaged ones showing fewer apoptotic cells and fewer changes in the stem cell signaling 24 h after irradiation, but displayed higher levels of chromosomal aberrations. Even though many of the observed changes were transient, surviving hES cells, which were differentiated 4 days postirradiation, showed a lower efficiency to form definitive endoderm than their mock-irradiated counterparts. This was demonstrated by lower expression levels of SOX17 and microRNA miR-375. In conclusion, hES cells are a suitable tool for the IR risk assessment during early human development. However, careful choice of the culture methods and a vigorous monitoring of the stem cell quality are mandatory for the use of these cells. Exposure to IR influences the stem cell properties of hES cells even when immediate radiation effects are overcome. This warrants consideration in the risk assessment of radiation effects during the earliest stages of human development.
Collapse
Affiliation(s)
- Sabine Luft
- 1 Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research , Darmstadt, Germany
| | - Onetsine Arrizabalaga
- 1 Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research , Darmstadt, Germany
| | - Ireen Kulish
- 1 Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research , Darmstadt, Germany .,2 Technical University Darmstadt , Darmstadt, Germany
| | - Elena Nasonova
- 1 Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research , Darmstadt, Germany .,3 Laboratory of Radiation Biology, Joint Institute for Nuclear Research , Dubna, Russia
| | - Marco Durante
- 1 Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research , Darmstadt, Germany
| | - Sylvia Ritter
- 1 Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research , Darmstadt, Germany
| | - Insa S Schroeder
- 1 Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research , Darmstadt, Germany
| |
Collapse
|
125
|
Bodenstine TM, Chandler GS, Seftor REB, Seftor EA, Hendrix MJC. Plasticity underlies tumor progression: role of Nodal signaling. Cancer Metastasis Rev 2016; 35:21-39. [PMID: 26951550 DOI: 10.1007/s10555-016-9605-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The transforming growth factor beta (TGFβ) superfamily member Nodal is an established regulator of early embryonic development, with primary roles in endoderm induction, left-right asymmetry, and primitive streak formation. Nodal signals through TGFβ family receptors at the plasma membrane and induces signaling cascades leading to diverse transcriptional regulation. While conceptually simple, the regulation of Nodal and its molecular effects are profoundly complex and context dependent. Pioneering work by developmental biologists has characterized the signaling pathways, regulatory components, and provided detailed insight into the mechanisms by which Nodal mediates changes at the cellular and organismal levels. Nodal is also an important factor in maintaining pluripotency of embryonic stem cells through regulation of core transcriptional programs. Collectively, this work has led to an appreciation for Nodal as a powerful morphogen capable of orchestrating multiple cellular phenotypes. Although Nodal is not active in most adult tissues, its reexpression and signaling have been linked to multiple types of human cancer, and Nodal has emerged as a driver of tumor growth and cellular plasticity. In vitro and in vivo experimental evidence has demonstrated that inhibition of Nodal signaling reduces cancer cell aggressive characteristics, while clinical data have established associations with Nodal expression and patient outcomes. As a result, there is great interest in the potential targeting of Nodal activity in a therapeutic setting for cancer patients that may provide new avenues for suppressing tumor growth and metastasis. In this review, we evaluate our current understanding of the complexities of Nodal function in cancer and highlight recent experimental evidence that sheds light on the therapeutic potential of its inhibition.
Collapse
Affiliation(s)
- Thomas M Bodenstine
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Grace S Chandler
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Richard E B Seftor
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Elisabeth A Seftor
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Mary J C Hendrix
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA.
| |
Collapse
|
126
|
Takasato M, Little MH. A strategy for generating kidney organoids: Recapitulating the development in human pluripotent stem cells. Dev Biol 2016; 420:210-220. [PMID: 27565022 PMCID: PMC6186756 DOI: 10.1016/j.ydbio.2016.08.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/19/2016] [Accepted: 08/21/2016] [Indexed: 02/06/2023]
Abstract
Directed differentiation of human pluripotent stem cells (hPSCs) can provide us any required tissue/cell types by recapitulating the development in vitro. The kidney is one of the most challenging organs to generate from hPSCs as the kidney progenitors are composed of at least 4 different cell types, including nephron, collecting duct, endothelial and interstitium progenitors, that are developmentally distinguished populations. Although the actual developmental process of the kidney during human embryogenesis has not been clarified yet, studies using model animals accumulated knowledge about the origins of kidney progenitors. The implications of these findings for the directed differentiation of hPSCs into the kidney include the mechanism of the intermediate mesoderm specification and its patterning along with anteroposterior axis. Using this knowledge, we previously reported successful generation of hPSCs-derived kidney organoids that contained all renal components and modelled human kidney development in vitro. In this review, we explain the developmental basis of the strategy behind this differentiation protocol and compare strategies of studies that also recently reported the induction of kidney cells from hPSCs. We also discuss the characterization of such kidney organoids and limitations and future applications of this technology.
Collapse
Affiliation(s)
- Minoru Takasato
- Murdoch Childrens Research Institute, Parkville, Victoria 3052, Australia; RIKEN Center for Developmental Biology, Kobe 650-0047, Japan.
| | - Melissa H Little
- Murdoch Childrens Research Institute, Parkville, Victoria 3052, Australia; Department of Pediatrics, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
127
|
Crespo D, Assis LHC, Furmanek T, Bogerd J, Schulz RW. Expression profiling identifies Sertoli and Leydig cell genes as Fsh targets in adult zebrafish testis. Mol Cell Endocrinol 2016; 437:237-251. [PMID: 27566230 DOI: 10.1016/j.mce.2016.08.033] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/27/2016] [Accepted: 08/22/2016] [Indexed: 11/26/2022]
Abstract
Spermatogonial stem cells are quiescent, undergo self-renewal or differentiating divisions, thereby forming the cellular basis of spermatogenesis. This cellular development is orchestrated by follicle-stimulating hormone (FSH), through the production of Sertoli cell-derived factors, and by Leydig cell-released androgens. Here, we investigate the transcriptional events induced by Fsh in a steroid-independent manner on the restart of zebrafish (Danio rerio) spermatogenesis ex vivo, using testis from adult males where type A spermatogonia were enriched by estrogen treatment in vivo. Under these conditions, RNA sequencing preferentially detected differentially expressed genes in somatic/Sertoli cells. Fsh-stimulated spermatogonial proliferation was accompanied by modulating several signaling systems (i.e. Tgf-β, Hedgehog, Wnt and Notch pathways). In silico protein-protein interaction analysis indicated a role for Hedgehog family members potentially integrating signals from different pathways during fish spermatogenesis. Moreover, Fsh had a marked impact on metabolic genes, such as lactate and fatty acid metabolism, or on Sertoli cell barrier components. Fish Leydig cells express the Fsh receptor and one of the most robust Fsh-responsive genes was insulin-like 3 (insl3), a Leydig cell-derived growth factor. Follow-up work showed that recombinant zebrafish Insl3 mediated pro-differentiation effects of Fsh on spermatogonia in an androgen-independent manner. Our experimental approach allowed focusing on testicular somatic genes in zebrafish and showed that the activity of signaling systems known to be relevant in stem cells was modulated by Fsh, providing promising leads for future work, as exemplified by the studies on Insl3.
Collapse
Affiliation(s)
- Diego Crespo
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Luiz H C Assis
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Tomasz Furmanek
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Jan Bogerd
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Rüdiger W Schulz
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands; Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway.
| |
Collapse
|
128
|
Development of a rapid screen for the endodermal differentiation potential of human pluripotent stem cell lines. Sci Rep 2016; 6:37178. [PMID: 27872482 PMCID: PMC5118706 DOI: 10.1038/srep37178] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 10/26/2016] [Indexed: 02/07/2023] Open
Abstract
A challenge facing the human pluripotent stem cell (hPSC) field is the variability observed in differentiation potential of hPSCs. Variability can lead to time consuming and costly optimisation to yield the cell type of interest. This is especially relevant for the differentiation of hPSCs towards the endodermal lineages. Endodermal cells have the potential to yield promising new knowledge and therapies for diseases affecting multiple organ systems, including lung, thymus, intestine, pancreas and liver, as well as applications in regenerative medicine and toxicology. Providing a means to rapidly, cheaply and efficiently assess the differentiation potential of multiple hPSCs is of great interest. To this end, we have developed a rapid small molecule based screen to assess the endodermal potential (EP) of hPSCs, based solely on definitive endoderm (DE) morphology. This drastically reduces the cost and time to identify lines suitable for use in deriving endodermal lineages. We demonstrate the efficacy of this screen using 10 different hPSCs, including 4 human embryonic stem cell lines (hESCs) and 6 human induced pluripotent stem cell lines (hiPSCs). The screen clearly revealed lines amenable to endodermal differentiation, and only lines that passed our morphological assessment were capable of further differentiation to hepatocyte like cells (HLCs).
Collapse
|
129
|
Conserved and divergent expression patterns of markers of axial development in the laboratory opossum,Monodelphis domestica. Dev Dyn 2016; 245:1176-1188. [DOI: 10.1002/dvdy.24459] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/14/2016] [Accepted: 09/14/2016] [Indexed: 11/07/2022] Open
|
130
|
Liu R, Wang JH, Xu C, Sun B, Kang SO. Activin pathway enhances colorectal cancer stem cell self-renew and tumor progression. Biochem Biophys Res Commun 2016; 479:715-720. [PMID: 27693580 DOI: 10.1016/j.bbrc.2016.09.146] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/28/2016] [Indexed: 12/28/2022]
Abstract
Activin belongs to transforming growth factor (TGF)-β super family of growth and differentiation factors and activin pathway participated in broad range of cell process. Studies elaborated activin pathway maintain pluripotency in human stem cells and suggest that the function of activin/nodal signaling in self-renew would be conserved across embryonic and adult stem cells. In this study, we tried to determine the effect of activin signaling pathway in regulation of cancer stem cells as a potential target for cancer therapy in clinical trials. A population of colorectal cancer cells was selected by the treatment of activin A. This population of cell possessed stem cell character with sphere formation ability. We demonstrated activin pathway enhanced the colorectal cancer stem cells self-renew and contribute to colorectal cancer progression in vivo. Targeting activin pathway potentially provide effective strategy for colorectal cancer therapy.
Collapse
Affiliation(s)
- Rui Liu
- Laboratory of Biophysics, School of Biological Sciences, Seoul National University, Seoul 151-742, Republic of Korea; Institute of Microbiology, Seoul National University, Seoul 151-742, Republic of Korea
| | - Jun-Hua Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Chengxiong Xu
- Cancer Center, Daping Hospital and Research Institute of Surgery, The Third Military Medical University, Chongqing 400042, China
| | - Bo Sun
- Institute of Microbiology, Seoul National University, Seoul 151-742, Republic of Korea; State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Sa-Ouk Kang
- Laboratory of Biophysics, School of Biological Sciences, Seoul National University, Seoul 151-742, Republic of Korea; Institute of Microbiology, Seoul National University, Seoul 151-742, Republic of Korea.
| |
Collapse
|
131
|
Kramer N, Rosner M, Kovacic B, Hengstschläger M. Full biological characterization of human pluripotent stem cells will open the door to translational research. Arch Toxicol 2016; 90:2173-2186. [PMID: 27325309 DOI: 10.1007/s00204-016-1763-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 06/13/2016] [Indexed: 12/13/2022]
Abstract
Since the discovery of human embryonic stem cells (hESC) and human-induced pluripotent stem cells (hiPSC), great hopes were held for their therapeutic application including disease modeling, drug discovery screenings, toxicological screenings and regenerative therapy. hESC and hiPSC have the advantage of indefinite self-renewal, thereby generating an inexhaustible pool of cells with, e.g., specific genotype for developing putative treatments; they can differentiate into derivatives of all three germ layers enabling autologous transplantation, and via donor-selection they can express various genotypes of interest for better disease modeling. Furthermore, drug screenings and toxicological screenings in hESC and hiPSC are more pertinent to identify drugs or chemical compounds that are harmful for human, than a mouse model could predict. Despite continuing research in the wide field of therapeutic applications, further understanding of the underlying basic mechanisms of stem cell function is necessary. Here, we summarize current knowledge concerning pluripotency, self-renewal, apoptosis, motility, epithelial-to-mesenchymal transition and differentiation of pluripotent stem cells.
Collapse
Affiliation(s)
- Nina Kramer
- Institute of Medical Genetics, Medical University of Vienna, Währingerstrasse 10, 1090, Vienna, Austria
| | - Margit Rosner
- Institute of Medical Genetics, Medical University of Vienna, Währingerstrasse 10, 1090, Vienna, Austria
| | - Boris Kovacic
- Institute of Medical Genetics, Medical University of Vienna, Währingerstrasse 10, 1090, Vienna, Austria
| | - Markus Hengstschläger
- Institute of Medical Genetics, Medical University of Vienna, Währingerstrasse 10, 1090, Vienna, Austria.
| |
Collapse
|
132
|
Pauklin S, Madrigal P, Bertero A, Vallier L. Initiation of stem cell differentiation involves cell cycle-dependent regulation of developmental genes by Cyclin D. Genes Dev 2016; 30:421-33. [PMID: 26883361 PMCID: PMC4762427 DOI: 10.1101/gad.271452.115] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Coordination of differentiation and cell cycle progression represents an essential process for embryonic development and adult tissue homeostasis. These mechanisms ultimately determine the quantities of specific cell types that are generated. Despite their importance, the precise molecular interplays between cell cycle machinery and master regulators of cell fate choice remain to be fully uncovered. Here, we demonstrate that cell cycle regulators Cyclin D1-3 control cell fate decisions in human pluripotent stem cells by recruiting transcriptional corepressors and coactivator complexes onto neuroectoderm, mesoderm, and endoderm genes. This activity results in blocking the core transcriptional network necessary for endoderm specification while promoting neuroectoderm factors. The genomic location of Cyclin Ds is determined by their interactions with the transcription factors SP1 and E2Fs, which result in the assembly of cell cycle-controlled transcriptional complexes. These results reveal how the cell cycle orchestrates transcriptional networks and epigenetic modifiers to instruct cell fate decisions.
Collapse
Affiliation(s)
- Siim Pauklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Madingley, Cambridge CB2 0SZ, United Kingdom
| | - Pedro Madrigal
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Madingley, Cambridge CB2 0SZ, United Kingdom; Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Alessandro Bertero
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Madingley, Cambridge CB2 0SZ, United Kingdom
| | - Ludovic Vallier
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Madingley, Cambridge CB2 0SZ, United Kingdom; Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| |
Collapse
|
133
|
Wang JQ, Liang WZ, Cui Y, He JT, Liu HY, Wang Y, Xue LX, Ji QY, Shi W, Shao YK, Mang J, Xu ZX. Noncanonical Activin A Signaling in PC12 Cells: A Self-Limiting Feedback Loop. Neurochem Res 2015; 41:1073-84. [DOI: 10.1007/s11064-015-1797-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 12/08/2015] [Accepted: 12/09/2015] [Indexed: 12/14/2022]
|
134
|
Li ASW, Marikawa Y. An in vitro gastrulation model recapitulates the morphogenetic impact of pharmacological inhibitors of developmental signaling pathways. Mol Reprod Dev 2015; 82:1015-36. [PMID: 26387793 DOI: 10.1002/mrd.22585] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 09/17/2015] [Indexed: 12/21/2022]
Abstract
Certain chemical agents act as teratogens, causing birth defects and fetal deaths when pregnant women are exposed to them. The establishment of in vitro models that recapitulate crucial embryonic events is therefore vital to facilitate screening of potential teratogens. Previously, we created a three-dimensional culture method for mouse P19C5 embryonal carcinoma stem cells that, when cultured as embryoid bodies, display elongation morphogenesis resembling gastrulation, which is the critical event resulting in the germ layers and major body axes. Determination of how well this in vitro morphogenesis represents in vivo gastrulation is essential to assess its applicability as well as to identify limitations of the model for detecting teratogenic agents. Here, we investigated the morphological and molecular characteristics of P19C5 morphogenesis using pharmacological agents that are known to cause abnormal patterning in the embryo in vivo by inhibiting major developmental signaling--e.g., involving Wnt, Nodal, Bone morphogenic protein (Bmp), Fibroblast growth factor (Fgf), Retinoic acid, Notch, and Hedgehog pathways. Inhibitors of Wnt, Nodal, Bmp, Fgf, and Retinoic acid signaling caused distinct changes in P19C5 morphogenesis that were quantifiable using morphometric parameters. These five inhibitors, plus the Notch inhibitor, also altered temporal expression profiles of developmental regulator genes in a manner consistent with the in vivo roles of the corresponding signaling pathways. In contrast, the Hedgehog inhibitor did not have any impact on the process, suggesting an absence of active Hedgehog signaling in these embryoid bodies. These results indicate that the P19C5 in vitro gastrulation model is a promising tool to screen for teratogenic agents that interfere with many of the key developmental signals.
Collapse
Affiliation(s)
- Aileen S W Li
- Developmental and Reproductive Biology Graduate Program, Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Hawaii
| | - Yusuke Marikawa
- Developmental and Reproductive Biology Graduate Program, Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Hawaii
| |
Collapse
|
135
|
Testa U, Castelli G, Elvira P. Experimental and investigational therapies for chemotherapy-induced anemia. Expert Opin Investig Drugs 2015; 24:1433-45. [PMID: 26359222 DOI: 10.1517/13543784.2015.1085505] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION In cancer patients, anemia is frequently observed, particularly as a consequence to chemotherapy (chemotherapy-induced anemia, CIA). CIA is treated with Red Blood Cell transfusions and erythropoiesis-stimulating agents (ESAs). However, the use of ESAs in anemic cancer patients is associated with reduced survival time and time to progression. Consequently, new therapeutic options are needed. AREAS COVERED In this article, the authors discuss new erythroid-enhancing agents (EEAs) that act differently to erythropoietin. Specifically, the article summarizes the early clinical development of activin antagonists (Sotatercep [ACE-011] and ACE-536) and hepcidin antagonists [NOX-H94]). EXPERT OPINION Both Activin RIIA trap agents and hepcidin inhibitors are promising new EEAs, but their safety profile, and their impact on treating CIA, needs to be carefully assessed in controlled clinical trials over longer periods of time. It is also important to carefully evaluate CIA patients to properly assess the physiopathological mechanisms responsible for the development of their anemic condition and provide patients with the most appropriate treatment plan.
Collapse
Affiliation(s)
- Ugo Testa
- a Istituto Superiore di Sanità, Department of Hematology, Oncology and Molecular Medicine , Viale Regina Elena 299, Rome, Italy
| | - Germana Castelli
- a Istituto Superiore di Sanità, Department of Hematology, Oncology and Molecular Medicine , Viale Regina Elena 299, Rome, Italy
| | - Pelosi Elvira
- a Istituto Superiore di Sanità, Department of Hematology, Oncology and Molecular Medicine , Viale Regina Elena 299, Rome, Italy
| |
Collapse
|
136
|
Abstract
Bone morphogenetic proteins (BMPs), together with the eponymous transforming growth factor (TGF) β and the Activins form the TGFβ superfamily of ligands. This protein family comprises more than 30 structurally highly related proteins, which determine formation, maintenance, and regeneration of tissues and organs. Their importance for the development of multicellular organisms is evident from their existence in all vertebrates as well as nonvertebrate animals. From their highly specific functions in vivo either a strict relation between a particular ligand and its cognate cellular receptor and/or a stringent regulation to define a distinct temperospatial expression pattern for the various ligands and receptor is expected. However, only a limited number of receptors are found to serve a large number of ligands thus implicating highly promiscuous ligand-receptor interactions instead. Since in tissues a multitude of ligands are often found, which signal via a highly overlapping set of receptors, this raises the question how such promiscuous interactions between different ligands and their receptors can generate concerted and highly specific cellular signals required during embryonic development and tissue homeostasis.
Collapse
Affiliation(s)
- Thomas D Mueller
- Department Plant Physiology and Biophysics, Julius-von-Sachs Institute of the University Wuerzburg, Wuerzburg, Germany.
| |
Collapse
|