1
|
Iqbal S, Andersson S, Nesta E, Pentinmikko N, Kumar A, Kumar Jha S, Borshagovski D, Webb A, Gebert N, Viitala EW, Ritchie A, Scharaw S, Kuuluvainen E, Larsen HL, Saarinen T, Juuti A, Ristimäki A, Jeltsch M, Ori A, Varjosalo M, Pietiläinen KH, Ollila S, Jensen KB, Oudhoff MJ, Katajisto P. Fetal-like reversion in the regenerating intestine is regulated by mesenchymal asporin. Cell Stem Cell 2025; 32:613-626.e8. [PMID: 40054463 DOI: 10.1016/j.stem.2025.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/03/2024] [Accepted: 02/12/2025] [Indexed: 04/06/2025]
Abstract
Mesenchymal cells and the extracellular matrix (ECM) support epithelium during homeostasis and regeneration. However, the role of the mesenchyme in epithelial conversion into a fetal-like regenerative state after damage is not known. We modeled epithelial regeneration by culturing intestinal epithelium on decellularized small intestinal scaffolds (iECM) and identify asporin (Aspn), an ECM-bound proteoglycan, as a critical mediator of epithelial fetal-like reprogramming. After damage, transient increase in Aspn expression by the pericryptal fibroblasts induces epithelial transforming growth factor β (TGF-β)-signaling via CD44 and promotes timely epithelial reprogramming. Temporal control of Aspn is lost in old mice, and after damage, the persistently high level of Aspn stagnates epithelium in the regenerative state. Increase in Wnt signaling can resolve the stagnated regenerative program of the old epithelium, promoting restoration of tissue function. In summary, we establish a platform for modeling epithelial injury responses ex vivo and show that the mesenchymal Aspn-producing niche modulates tissue repair by regulating epithelial fetal-like reprogramming.
Collapse
Affiliation(s)
- Sharif Iqbal
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland; Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Simon Andersson
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland; Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Ernesta Nesta
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Nalle Pentinmikko
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ashish Kumar
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sawan Kumar Jha
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Daniel Borshagovski
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Anna Webb
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Nadja Gebert
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Emma W Viitala
- Translational Cancer Medicine Program, University of Helsinki, Helsinki, Finland
| | - Alexandra Ritchie
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland; Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Sandra Scharaw
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Emilia Kuuluvainen
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland; Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Hjalte L Larsen
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Tuure Saarinen
- Helsinki University Hospital, Abdominal Center, Department of Endocrinology, Obesity Center, Helsinki, Finland
| | - Anne Juuti
- Helsinki University Hospital, Abdominal Center, Department of Endocrinology, Obesity Center, Helsinki, Finland
| | - Ari Ristimäki
- Department of Pathology, HUSLAB, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00290 Helsinki, Finland; Applied Tumor Genomics Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, 00140 Helsinki, Finland
| | - Michael Jeltsch
- Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Alessandro Ori
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Markku Varjosalo
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kirsi H Pietiläinen
- Helsinki University Hospital, Abdominal Center, Department of Endocrinology, Obesity Center, Helsinki, Finland; Obesity Research Unit, Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Saara Ollila
- Translational Cancer Medicine Program, University of Helsinki, Helsinki, Finland
| | - Kim B Jensen
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Menno J Oudhoff
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway; Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| | - Pekka Katajisto
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland; Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland; Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
2
|
Feng C, Fan H, Tie R, Xin S, Chen M. Deciphering the evolving niche interactome of human hematopoietic stem cells from ontogeny to aging. Front Mol Biosci 2024; 11:1479605. [PMID: 39698109 PMCID: PMC11652281 DOI: 10.3389/fmolb.2024.1479605] [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: 08/12/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
Hematopoietic stem cells (HSC) reside within specialized microenvironments that undergo dynamic changes throughout development and aging to support HSC function. However, the evolving cell-cell communication networks within these niches remain largely unexplored. This study integrates single-cell RNA sequencing datasets to systematically characterize the HSC niche interactome from ontogeny to aging. We reconstructed single-cell atlases of HSC niches at different developmental stages, revealing stage-specific cellular compositions and interactions targeting HSC. During HSC maturation, our analysis identified distinct patterns of ligand-receptor interactions and signaling pathways that govern HSC emergence, expansion, and maintenance. HSC aging was accompanied by a decrease in supportive niche interactions, followed by an adaptive increase in interaction strength in old adult bone marrow. This complex aging process involved the emergence of interactions associated with inflammation, altered stem cell function, and a decline in the efficacy of key signaling pathways. Our findings provide a comprehensive understanding of the dynamic remodeling of the HSC niche interactome throughout life, paving the way for targeted interventions to maintain HSC function and promote healthy aging. This study offers valuable insights into the intricate cell-cell communication networks that govern HSC behavior and fate, with implications for hematological disorders and regenerative medicine.
Collapse
Affiliation(s)
- Cong Feng
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, China
- Bioinformatics Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haoyan Fan
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, China
| | - Ruxiu Tie
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Hematology-Oncology, Taizhou Hospital of Zhejiang Province, Linhai, China
| | - Saige Xin
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Ming Chen
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, China
- Bioinformatics Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Elahimanesh M, Shokri N, Mohammadi P, Parvaz N, Najafi M. Step by step analysis on gene datasets of growth phases in hematopoietic stem cells. Biochem Biophys Rep 2024; 39:101737. [PMID: 38881758 PMCID: PMC11176649 DOI: 10.1016/j.bbrep.2024.101737] [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: 01/24/2024] [Revised: 05/16/2024] [Accepted: 05/19/2024] [Indexed: 06/18/2024] Open
Abstract
Background Umbilical cord blood hematopoietic stem cells (UCB-HSCs) have important roles in the treatment of illnesses based on their self-renewal and potency characteristics. Knowing the gene profiles and signaling pathways involved in each step of the cell cycle could improve the therapeutic approaches of HSCs. The aim of this study was to predict the gene profiles and signaling pathways involved in the G0, G1, and differentiation stages of HSCs. Methods Interventional (n = 8) and non-interventional (n = 3) datasets were obtained from the Gene Expression Omnibus (GEO) database, and were crossed and analyzed to determine the high- and low-express genes related to each of the G0, G1, and differentiation stages of HSCs. Then, the scores of STRING were annotated to the gene data. The gene networks were constructed using Cytoscape software, and enriched with the KEGG and GO databases. Results The high- and low-express genes were determined due to inter and intra intersections of the interventional and non-interventional data. The non-interventional data were applied to construct the gene networks (n = 6) with the nodes improved using the interventional data. Several important signaling pathways were suggested in each of the G0, G1, and differentiation stages. Conclusion The data revealed that the different signaling pathways are activated in each of the G0, G1, and differentiation stages so that their genes may be targeted to improve the HSC therapy.
Collapse
Affiliation(s)
- Mohammad Elahimanesh
- Clinical Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Shokri
- Clinical Biochemistry Department, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Payam Mohammadi
- Clinical Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Najmeh Parvaz
- Clinical Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Clinical Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Laowtammathron C, Lorthongpanich C, Jiamvoraphong N, Srisook P, Klaihmon P, Kheolamai P, Luanpitpong S, Issaragrisil S. Role of YAP in hematopoietic differentiation and erythroid lineage specification of human-induced pluripotent stem cells. Stem Cell Res Ther 2023; 14:279. [PMID: 37775798 PMCID: PMC10543272 DOI: 10.1186/s13287-023-03508-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 09/21/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND In vitro production of hematopoietic stem/progenitor cells (HSPCs) from human-induced pluripotent stem cells (hiPSCs) provides opportunities for fundamental research, disease modeling, and large-scale production of HLA-matched HSPCs for therapeutic applications. However, a comprehensive understanding of the signaling mechanisms that regulate human hematopoiesis is needed to develop a more effective procedure for deriving HSPCs from hiPSCs. METHODS In this study, we investigate the role of YAP during the hematopoietic differentiation of hiPSCs to HSPCs and erythrocytes using the isogenic YAP-overexpressing (YAP-S5A) and YAP-depleting (YAP-KD) hiPSCs to eliminate the effects of a genetic background variation. RESULTS Although YAP is dispensable for maintaining the self-renewal and pluripotency of these hiPSCs, it affects the early cell-fate determination and hematopoietic differentiation of hiPSCs. Depleting YAP enhances the derivation efficiency of HSPCs from hiPSCs by inducing the mesodermal lineage commitment, promoting hematopoietic differentiation, and preventing the differentiation toward endothelial lineage. On the contrary, the overexpression of YAP reduced HSPCs yield by inducing the endodermal lineage commitment, suppressing hematopoietic differentiation, and promoting the differentiation toward endothelial lineage. CONCLUSIONS Expression of YAP is crucial for the differentiation of hiPSC-derived HSPCs toward mature erythrocytes. We believe that by manipulating YAP activity using small molecules, the efficiency of the large-scale in vitro production system for generating hematopoietic stem/progenitor cells for future therapeutic use could be improved.
Collapse
Affiliation(s)
- Chuti Laowtammathron
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Chanchao Lorthongpanich
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| | - Nittaya Jiamvoraphong
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Pimonwan Srisook
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Phatchanat Klaihmon
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Pakpoom Kheolamai
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Bangkok Hematology Center, Wattanosoth Hospital, BDMS Center of Excellence for Cancer, Bangkok, Thailand
| |
Collapse
|
5
|
Kim I, Park T, Noh JY, Kim W. Emerging role of Hippo pathway in the regulation of hematopoiesis. BMB Rep 2023; 56:417-425. [PMID: 37574808 PMCID: PMC10471462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/27/2023] [Accepted: 07/28/2023] [Indexed: 08/15/2023] Open
Abstract
In various organisms, the Hippo signaling pathway has been identified as a master regulator of organ size determination and tissue homeostasis. The Hippo signaling coordinates embryonic development, tissue regeneration and differentiation, through regulating cell proliferation and survival. The YAP and TAZ (YAP/TAZ) act as core transducers of the Hippo pathway, and they are tightly and exquisitely regulated in response to various intrinsic and extrinsic stimuli. Abnormal regulation or genetic variation of the Hippo pathway causes a wide range of human diseases, including cancer. Recent studies have revealed that Hippo signaling plays a pivotal role in the immune system and cancer immunity. Due to pathophysiological importance, the emerging role of Hippo signaling in blood cell differentiation, known as hematopoiesis, is receiving much attention. A number of elegant studies using a genetically engineered mouse (GEM) model have shed light on the mechanistic and physiological insights into the Hippo pathway in the regulation of hematopoiesis. Here, we briefly review the function of Hippo signaling in the regulation of hematopoiesis and immune cell differentiation. [BMB Reports 2023; 56(8): 417-425].
Collapse
Affiliation(s)
- Inyoung Kim
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Korea
| | - Taeho Park
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon 34113, Korea
| | - Ji-Yoon Noh
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon 34113, Korea
| | - Wantae Kim
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
6
|
Pikkupeura LM, Bressan RB, Guiu J, Chen Y, Maimets M, Mayer D, Schweiger PJ, Hansen SL, Maciag GJ, Larsen HL, Lõhmussaar K, Pedersen MT, Teves JMY, Bornholdt J, Benes V, Sandelin A, Jensen KB. Transcriptional and epigenomic profiling identifies YAP signaling as a key regulator of intestinal epithelium maturation. SCIENCE ADVANCES 2023; 9:eadf9460. [PMID: 37436997 DOI: 10.1126/sciadv.adf9460] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 06/08/2023] [Indexed: 07/14/2023]
Abstract
During intestinal organogenesis, equipotent epithelial progenitors mature into phenotypically distinct stem cells that are responsible for lifelong maintenance of the tissue. While the morphological changes associated with the transition are well characterized, the molecular mechanisms underpinning the maturation process are not fully understood. Here, we leverage intestinal organoid cultures to profile transcriptional, chromatin accessibility, DNA methylation, and three-dimensional (3D) chromatin conformation landscapes in fetal and adult epithelial cells. We observed prominent differences in gene expression and enhancer activity, which are accompanied by local changes in 3D organization, DNA accessibility, and methylation between the two cellular states. Using integrative analyses, we identified sustained Yes-Associated Protein (YAP) transcriptional activity as a major gatekeeper of the immature fetal state. We found the YAP-associated transcriptional network to be regulated at various levels of chromatin organization and likely to be coordinated by changes in extracellular matrix composition. Together, our work highlights the value of unbiased profiling of regulatory landscapes for the identification of key mechanisms underlying tissue maturation.
Collapse
Affiliation(s)
- Laura M Pikkupeura
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N DK-2200, Denmark
- Bioinformatics Center, Department of Biology, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Raul B Bressan
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N DK-2200, Denmark
- Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Jordi Guiu
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N DK-2200, Denmark
- Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, 3a planta, Av. Granvia de l'Hospitalet 199, Hospitalet de Llobregat 08908, Spain
| | - Yun Chen
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N DK-2200, Denmark
- Bioinformatics Center, Department of Biology, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Martti Maimets
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N DK-2200, Denmark
- Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Daniela Mayer
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N DK-2200, Denmark
- Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Pawel J Schweiger
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N DK-2200, Denmark
- Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Stine L Hansen
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N DK-2200, Denmark
- Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Grzegorz J Maciag
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N DK-2200, Denmark
- Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Hjalte L Larsen
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N DK-2200, Denmark
- Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Kadi Lõhmussaar
- Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | | | - Joji M Yap Teves
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N DK-2200, Denmark
- Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Jette Bornholdt
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N DK-2200, Denmark
- Bioinformatics Center, Department of Biology, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | | | - Albin Sandelin
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N DK-2200, Denmark
- Bioinformatics Center, Department of Biology, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Kim B Jensen
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N DK-2200, Denmark
- Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N DK-2200, Denmark
| |
Collapse
|
7
|
Meli VS, Veerasubramanian PK, Downing TL, Wang W, Liu WF. Mechanosensation to inflammation: Roles for YAP/TAZ in innate immune cells. Sci Signal 2023; 16:eadc9656. [PMID: 37130167 PMCID: PMC10625748 DOI: 10.1126/scisignal.adc9656] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 04/14/2023] [Indexed: 05/04/2023]
Abstract
Innate immune cells are responsible for eliminating foreign infectious agents and cellular debris, and their ability to perceive, respond to, and integrate biochemical and mechanical cues from their microenvironment eventually determines their behavior. In response to tissue injury, pathogen invasion, or a biomaterial implant, immune cells activate many pathways to initiate inflammation in the tissue. In addition to common inflammatory pathways, studies have demonstrated the role of the mechanosensitive proteins and transcriptional coactivators YAP and TAZ (YAP/TAZ) in inflammation and immunity. We review our knowledge of YAP/TAZ in controlling inflammation and immunity in innate immune cells. Furthermore, we discuss the roles of YAP/TAZ in inflammatory diseases, wound healing, and tissue regeneration and how they integrate mechanical cues with biochemical signaling during disease progression. Last, we comment on possible approaches that can be exploited to harness the therapeutic potential of YAP/TAZ in inflammatory diseases.
Collapse
Affiliation(s)
- Vijaykumar S. Meli
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, (CIRC), University of California Irvine, CA 92697
| | - Praveen Krishna Veerasubramanian
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, (CIRC), University of California Irvine, CA 92697
| | - Timothy L. Downing
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, (CIRC), University of California Irvine, CA 92697
- NSF-Simons Center for Multiscale Cell Fate Research, University of California Irvine, CA 92697
- Department of Microbiology and Molecular Genetics, University of California Irvine, CA 92697
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California Irvine, CA 92697
| | - Wendy F. Liu
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, (CIRC), University of California Irvine, CA 92697
- Department of Chemical and Biomolecular Engineering, University of California Irvine, CA 92697
- Department of Molecular Biology and Biochemistry, University of California Irvine, CA 92697
- Institute for Immunology, University of California Irvine, CA 92697
| |
Collapse
|
8
|
Abstract
Immune responses are governed by signals from the tissue microenvironment, and in addition to biochemical signals, mechanical cues and forces arising from the tissue, its extracellular matrix and its constituent cells shape immune cell function. Indeed, changes in biophysical properties of tissue alter the mechanical signals experienced by cells in many disease conditions, in inflammatory states and in the context of ageing. These mechanical cues are converted into biochemical signals through the process of mechanotransduction, and multiple pathways of mechanotransduction have been identified in immune cells. Such pathways impact important cellular functions including cell activation, cytokine production, metabolism, proliferation and trafficking. Changes in tissue mechanics may also represent a new form of 'danger signal' that alerts the innate and adaptive immune systems to the possibility of injury or infection. Tissue mechanics can change temporally during an infection or inflammatory response, offering a novel layer of dynamic immune regulation. Here, we review the emerging field of mechanoimmunology, focusing on how mechanical cues at the scale of the tissue environment regulate immune cell behaviours to initiate, propagate and resolve the immune response.
Collapse
|
9
|
Damkham N, Issaragrisil S, Lorthongpanich C. Role of YAP as a Mechanosensing Molecule in Stem Cells and Stem Cell-Derived Hematopoietic Cells. Int J Mol Sci 2022; 23:14634. [PMID: 36498961 PMCID: PMC9737411 DOI: 10.3390/ijms232314634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/11/2022] [Accepted: 11/20/2022] [Indexed: 11/25/2022] Open
Abstract
Yes-associated protein (YAP) and WW domain-containing transcription regulator protein 1 (WWTR1, also known as TAZ) are transcriptional coactivators in the Hippo signaling pathway. Both are well-known regulators of cell proliferation and organ size control, and they have significant roles in promoting cell proliferation and differentiation. The roles of YAP and TAZ in stem cell pluripotency and differentiation have been extensively studied. However, the upstream mediators of YAP and TAZ are not well understood. Recently, a novel role of YAP in mechanosensing and mechanotransduction has been reported. The present review updates information on the regulation of YAP by mechanical cues such as extracellular matrix stiffness, fluid shear stress, and actin cytoskeleton tension in stem cell behaviors and differentiation. The review explores mesenchymal stem cell fate decisions, pluripotent stem cells (PSCs), self-renewal, pluripotency, and differentiation to blood products. Understanding how cells sense their microenvironment or niche and mimic those microenvironments in vitro could improve the efficiency of producing stem cell products and the efficacy of the products.
Collapse
Affiliation(s)
- Nattaya Damkham
- Siriraj Center of Excellence for Stem cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Bangkok Hematology Center, Wattanosoth Hospital, BDMS Center of Excellence for Cancer, Bangkok 10310, Thailand
| | - Chanchao Lorthongpanich
- Siriraj Center of Excellence for Stem cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
10
|
Damkham N, Lorthongpanich C, Klaihmon P, Lueangamornnara U, Kheolamai P, Trakarnsanga K, Issaragrisil S. YAP and TAZ play a crucial role in human erythrocyte maturation and enucleation. Stem Cell Res Ther 2022; 13:467. [PMID: 36076260 PMCID: PMC9461202 DOI: 10.1186/s13287-022-03166-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Yes-associated protein (YAP) and WW domain-containing transcription regulator protein 1 (WWTR1, also known as TAZ) are two key transcription co-activators of the Hippo pathway. Both were originally characterized as organ size and cell proliferation regulators. Later studies demonstrated that the Hippo pathway may play a role in Drosophila and mammal hematopoiesis. However, the role of the Hippo pathway in human erythropoiesis has not yet been fully elucidated. METHODS The role of YAP and TAZ was studied in human erythropoiesis and hematopoietic stem cell (HSC) lineage determination by using mobilized peripheral blood (PB) and cord blood (CB)-derived HSC as a model. HSCs were isolated and cultured in an erythroid differentiation medium for erythroid differentiation and culture in methylcellulose assay for HSC lineage determination study. RESULTS YAP and TAZ were barely detectable in human HSCs, but became highly expressed in pro-erythroblasts and erythroblasts. Depletion or knockdown of YAP and/or TAZ did not affect the ability of HSC lineage specification to erythroid lineage in either methylcellulose assay or liquid culture. However, depletion of YAP and TAZ did impair erythroblast terminal differentiation to erythrocytes and their enucleation. Moreover, ectopic expression of YAP and TAZ in pro-erythroblasts did not exert an apparent effect on erythroid differentiation, expansion, or morphology. CONCLUSIONS This study demonstrated that YAP/TAZ plays important role in erythroid maturation and enucleation but is dispensable for lineage determination of human HSCs.
Collapse
Affiliation(s)
- Nattaya Damkham
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
| | - Chanchao Lorthongpanich
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Siriraj, Bangkoknoi, Bangkok, 10700, Thailand.
| | - Phatchanat Klaihmon
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
| | - Usaneeporn Lueangamornnara
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pakpoom Kheolamai
- Division of Cell Biology, Department of Pre-Clinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Kongtana Trakarnsanga
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Siriraj, Bangkoknoi, Bangkok, 10700, Thailand.
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
- Bangkok Hematology Center, Wattanosoth Hospital, BDMS Center of Excellence for Cancer, Bangkok, Thailand.
| |
Collapse
|
11
|
Fernández-García V, González-Ramos S, Martín-Sanz P, Castrillo A, Boscá L. Unraveling the interplay between iron homeostasis, ferroptosis and extramedullary hematopoiesis. Pharmacol Res 2022; 183:106386. [PMID: 35933006 DOI: 10.1016/j.phrs.2022.106386] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/25/2022] [Accepted: 08/02/2022] [Indexed: 11/25/2022]
Abstract
Iron participates in myriad processes necessary to sustain life. During the past decades, great efforts have been made to understand iron regulation and function in health and disease. Indeed, iron is associated with both physiological (e.g., immune cell biology and function and hematopoiesis) and pathological (e.g., inflammatory and infectious diseases, ferroptosis and ferritinophagy) processes, yet few studies have addressed the potential functional link between iron, the aforementioned processes and extramedullary hematopoiesis, despite the obvious benefits that this could bring to clinical practice. Further investigation in this direction will shape the future development of individualized treatments for iron-linked diseases and chronic inflammatory disorders, including extramedullary hematopoiesis, metabolic syndrome, cardiovascular diseases and cancer.
Collapse
Affiliation(s)
- Victoria Fernández-García
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Universidad Autónoma de Madrid, Madrid, Spain.
| | - Silvia González-Ramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain.
| |
Collapse
|
12
|
Hippo pathway-related genes expression is deregulated in myeloproliferative neoplasms. Med Oncol 2022; 39:97. [DOI: 10.1007/s12032-022-01696-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/22/2022] [Indexed: 10/18/2022]
|
13
|
Sugden WW, North TE. Making Blood from the Vessel: Extrinsic and Environmental Cues Guiding the Endothelial-to-Hematopoietic Transition. Life (Basel) 2021; 11:life11101027. [PMID: 34685398 PMCID: PMC8539454 DOI: 10.3390/life11101027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 01/10/2023] Open
Abstract
It is increasingly recognized that specialized subsets of endothelial cells carry out unique functions in specific organs and regions of the vascular tree. Perhaps the most striking example of this specialization is the ability to contribute to the generation of the blood system, in which a distinct population of “hemogenic” endothelial cells in the embryo transforms irreversibly into hematopoietic stem and progenitor cells that produce circulating erythroid, myeloid and lymphoid cells for the lifetime of an animal. This review will focus on recent advances made in the zebrafish model organism uncovering the extrinsic and environmental factors that facilitate hemogenic commitment and the process of endothelial-to-hematopoietic transition that produces blood stem cells. We highlight in particular biomechanical influences of hemodynamic forces and the extracellular matrix, metabolic and sterile inflammatory cues present during this developmental stage, and outline new avenues opened by transcriptomic-based approaches to decipher cell–cell communication mechanisms as examples of key signals in the embryonic niche that regulate hematopoiesis.
Collapse
Affiliation(s)
- Wade W. Sugden
- Stem Cell Program, Department of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA;
- Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA
| | - Trista E. North
- Stem Cell Program, Department of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA;
- Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA
- Correspondence:
| |
Collapse
|
14
|
Yap1-Scribble polarization is required for hematopoietic stem cell division and fate. Blood 2021; 136:1824-1836. [PMID: 32483624 DOI: 10.1182/blood.2019004113] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
Yap1 and its paralogue Taz largely control epithelial tissue growth. We have identified that hematopoietic stem cell (HSC) fitness response to stress depends on Yap1 and Taz. Deletion of Yap1 and Taz induces a loss of HSC quiescence, symmetric self-renewal ability, and renders HSC more vulnerable to serial myeloablative 5-fluorouracil treatment. This effect depends on the predominant cytosolic polarization of Yap1 through a PDZ domain-mediated interaction with the scaffold Scribble. Scribble and Yap1 coordinate to control cytoplasmic Cdc42 activity and HSC fate determination in vivo. Deletion of Scribble disrupts Yap1 copolarization with Cdc42 and decreases Cdc42 activity, resulting in increased self-renewing HSC with competitive reconstitution advantages. These data suggest that Scribble/Yap1 copolarization is indispensable for Cdc42-dependent activity on HSC asymmetric division and fate. The combined loss of Scribble, Yap1, and Taz results in transcriptional upregulation of Rac-specific guanine nucleotide exchange factors, Rac activation, and HSC fitness restoration. Scribble links Cdc42 and the cytosolic functions of the Hippo signaling cascade in HSC fate determination.
Collapse
|
15
|
McGinn J, Hallou A, Han S, Krizic K, Ulyanchenko S, Iglesias-Bartolome R, England FJ, Verstreken C, Chalut KJ, Jensen KB, Simons BD, Alcolea MP. A biomechanical switch regulates the transition towards homeostasis in oesophageal epithelium. Nat Cell Biol 2021; 23:511-525. [PMID: 33972733 PMCID: PMC7611004 DOI: 10.1038/s41556-021-00679-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/01/2021] [Indexed: 02/07/2023]
Abstract
Epithelial cells rapidly adapt their behaviour in response to increasing tissue demands. However, the processes that finely control these cell decisions remain largely unknown. The postnatal period covering the transition between early tissue expansion and the establishment of adult homeostasis provides a convenient model with which to explore this question. Here, we demonstrate that the onset of homeostasis in the epithelium of the mouse oesophagus is guided by the progressive build-up of mechanical strain at the organ level. Single-cell RNA sequencing and whole-organ stretching experiments revealed that the mechanical stress experienced by the growing oesophagus triggers the emergence of a bright Krüppel-like factor 4 (KLF4) committed basal population, which balances cell proliferation and marks the transition towards homeostasis in a yes-associated protein (YAP)-dependent manner. Our results point to a simple mechanism whereby mechanical changes experienced at the whole-tissue level are integrated with those sensed at the cellular level to control epithelial cell fate.
Collapse
Affiliation(s)
- Jamie McGinn
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Oncology, University of Cambridge and Cancer Research UK Cambridge Centre, Cambridge, UK
| | - Adrien Hallou
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Seungmin Han
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - Kata Krizic
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Svetlana Ulyanchenko
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ramiro Iglesias-Bartolome
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Frances J England
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | | | - Kevin J Chalut
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Kim B Jensen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Benjamin D Simons
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge, UK
| | - Maria P Alcolea
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Oncology, University of Cambridge and Cancer Research UK Cambridge Centre, Cambridge, UK.
| |
Collapse
|
16
|
Krishnan M, Kumar S, Kangale LJ, Ghigo E, Abnave P. The Act of Controlling Adult Stem Cell Dynamics: Insights from Animal Models. Biomolecules 2021; 11:biom11050667. [PMID: 33946143 PMCID: PMC8144950 DOI: 10.3390/biom11050667] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/02/2021] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Adult stem cells (ASCs) are the undifferentiated cells that possess self-renewal and differentiation abilities. They are present in all major organ systems of the body and are uniquely reserved there during development for tissue maintenance during homeostasis, injury, and infection. They do so by promptly modulating the dynamics of proliferation, differentiation, survival, and migration. Any imbalance in these processes may result in regeneration failure or developing cancer. Hence, the dynamics of these various behaviors of ASCs need to always be precisely controlled. Several genetic and epigenetic factors have been demonstrated to be involved in tightly regulating the proliferation, differentiation, and self-renewal of ASCs. Understanding these mechanisms is of great importance, given the role of stem cells in regenerative medicine. Investigations on various animal models have played a significant part in enriching our knowledge and giving In Vivo in-sight into such ASCs regulatory mechanisms. In this review, we have discussed the recent In Vivo studies demonstrating the role of various genetic factors in regulating dynamics of different ASCs viz. intestinal stem cells (ISCs), neural stem cells (NSCs), hematopoietic stem cells (HSCs), and epidermal stem cells (Ep-SCs).
Collapse
Affiliation(s)
- Meera Krishnan
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Gurgaon-Faridabad Ex-pressway, Faridabad 121001, India; (M.K.); (S.K.)
| | - Sahil Kumar
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Gurgaon-Faridabad Ex-pressway, Faridabad 121001, India; (M.K.); (S.K.)
| | - Luis Johnson Kangale
- IRD, AP-HM, SSA, VITROME, Aix-Marseille University, 13385 Marseille, France;
- Institut Hospitalo Universitaire Méditerranée Infection, 13385 Marseille, France;
| | - Eric Ghigo
- Institut Hospitalo Universitaire Méditerranée Infection, 13385 Marseille, France;
- TechnoJouvence, 13385 Marseille, France
| | - Prasad Abnave
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Gurgaon-Faridabad Ex-pressway, Faridabad 121001, India; (M.K.); (S.K.)
- Correspondence:
| |
Collapse
|
17
|
Meli VS, Atcha H, Veerasubramanian PK, Nagalla RR, Luu TU, Chen EY, Guerrero-Juarez CF, Yamaga K, Pandori W, Hsieh JY, Downing TL, Fruman DA, Lodoen MB, Plikus MV, Wang W, Liu WF. YAP-mediated mechanotransduction tunes the macrophage inflammatory response. SCIENCE ADVANCES 2020; 6:eabb8471. [PMID: 33277245 PMCID: PMC7717914 DOI: 10.1126/sciadv.abb8471] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 10/20/2020] [Indexed: 05/11/2023]
Abstract
Macrophages are innate immune cells that adhere to the extracellular matrix within tissues. However, how matrix properties regulate their function remains poorly understood. Here, we report that the adhesive microenvironment tunes the macrophage inflammatory response through the transcriptional coactivator YAP. We find that adhesion to soft hydrogels reduces inflammation when compared to adhesion on stiff materials and is associated with reduced YAP expression and nuclear localization. Substrate stiffness and cytoskeletal polymerization, but not adhesive confinement nor contractility, regulate YAP localization. Furthermore, depletion of YAP inhibits macrophage inflammation, whereas overexpression of active YAP increases inflammation. Last, we show in vivo that soft materials reduce expression of inflammatory markers and YAP in surrounding macrophages when compared to stiff materials. Together, our studies identify YAP as a key molecule for controlling inflammation and sensing stiffness in macrophages and may have broad implications in the regulation of macrophages in health and disease.
Collapse
Affiliation(s)
- Vijaykumar S Meli
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
| | - Hamza Atcha
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
| | - Praveen Krishna Veerasubramanian
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
| | - Raji R Nagalla
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
| | - Thuy U Luu
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Esther Y Chen
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, CA, USA
| | - Christian F Guerrero-Juarez
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Kosuke Yamaga
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - William Pandori
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA
| | - Jessica Y Hsieh
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
| | - Timothy L Downing
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA
| | - David A Fruman
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA
| | - Melissa B Lodoen
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Wendy F Liu
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA.
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, CA, USA
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
18
|
A single copy of large tumor suppressor 1 or large tumor suppressor 2 is sufficient for normal hematopoiesis. Chin Med J (Engl) 2020; 133:1943-1951. [PMID: 32826458 PMCID: PMC7462215 DOI: 10.1097/cm9.0000000000000934] [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] [Indexed: 11/26/2022] Open
Abstract
Background Hematopoietic stem cells (HSCs) have the ability to differentiate into all subsets of blood cells and self-renew. Large tumor suppressor 1 (LATS1) and large tumor suppressor 2 (LATS2) kinases are essential for cell cycle regulation, organism fitness, genome integrity, and cancer prevention. Here, we investigated whether Lats1 and Lats2 are critical for the maintenance of the self-renewal and quiescence capacities of HSCs in mice. Methods Quantitative reverse transcription-polymerase chain reaction was used to determine the expression levels of Lats1 and Lats2 in subsets of progenitor cells and mature bone marrow cells. A clustered regularly interspaced short palindromic repeats system was used to generate Lats1 or Lats2 knockout mice. Complete blood cell counts were used to compare the absolute number of white blood cells, lymphocytes, monocytes, neutrophils, and platelets between Lats1 or Lats2 heterozygotes and littermates. Flow cytometry was used to assess the size of hematopoietic progenitor cells (HPCs) and HSC pools in Lats1 or Lats2 heterozygotes and littermates. The comparison between the two groups was analyzed using Student's t test. Results Lats1 and Lats2 were widely expressed in hematopoietic cells with higher expression levels in primitive hematopoietic cells than in mature cells. Lats1 or Lats2 knockout mice were generated, with the homozygotes showing embryonic lethality. The size of the HPC and HSC pools in Lats1 (HPC: wild-type [WT] vs. heterozygote, 220,426.77 ± 54,384.796 vs. 221,149.4 ± 42,688.29, P = 0.988; HSC: WT vs. heterozygote, 2498.932 ± 347.856 vs. 3249.763 ± 370.412, P = 0.105) or Lats2 (HPC: WT vs. heterozygote, 425,540.52 ± 99,721.86 vs. 467,127.8 ± 89,574.48, P = 0.527; HSC: WT vs. heterozygote, 4760.545 ± 1518.01 vs. 5327.437 ± 873.297, P = 0.502) heterozygotes were not impaired. Moreover, the depletion of Lats1 or Lats2 did not affect the overall survival of the heterozygotes (Lats1: P = 0.654; Lats2: P = 0.152). Conclusion These results indicate that a single allele of Lats1 or Lats2 may be sufficient for normal hematopoiesis.
Collapse
|
19
|
Lundin V, Sugden WW, Theodore LN, Sousa PM, Han A, Chou S, Wrighton PJ, Cox AG, Ingber DE, Goessling W, Daley GQ, North TE. YAP Regulates Hematopoietic Stem Cell Formation in Response to the Biomechanical Forces of Blood Flow. Dev Cell 2020; 52:446-460.e5. [PMID: 32032546 DOI: 10.1016/j.devcel.2020.01.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 10/16/2019] [Accepted: 01/07/2020] [Indexed: 12/27/2022]
Abstract
Hematopoietic stem and progenitor cells (HSPCs), first specified from hemogenic endothelium (HE) in the ventral dorsal aorta (VDA), support lifelong hematopoiesis. Their de novo production promises significant therapeutic value; however, current in vitro approaches cannot efficiently generate multipotent long-lived HSPCs. Presuming this reflects a lack of extrinsic cues normally impacting the VDA, we devised a human dorsal aorta-on-a-chip platform that identified Yes-activated protein (YAP) as a cyclic stretch-induced regulator of HSPC formation. In the zebrafish VDA, inducible Yap overexpression significantly increased runx1 expression in vivo and the number of CD41+ HSPCs downstream of HE specification. Endogenous Yap activation by lats1/2 knockdown or Rho-GTPase stimulation mimicked Yap overexpression and induced HSPCs in embryos lacking blood flow. Notably, in static human induced pluripotent stem cell (iPSC)-derived HE culture, compound-mediated YAP activation enhanced RUNX1 levels and hematopoietic colony-forming potential. Together, our findings reveal a potent impact of hemodynamic Rho-YAP mechanotransduction on HE fate, relevant to de novo human HSPC production.
Collapse
Affiliation(s)
- Vanessa Lundin
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Wade W Sugden
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Lindsay N Theodore
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Patricia M Sousa
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Areum Han
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Stephanie Chou
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Paul J Wrighton
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew G Cox
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Wolfram Goessling
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA 02115, USA
| | - George Q Daley
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Trista E North
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
20
|
Hao S, Matsui Y, Lai ZC, Paulson RF. Yap1 promotes proliferation of transiently amplifying stress erythroid progenitors during erythroid regeneration. Exp Hematol 2019; 80:42-54.e4. [PMID: 31756359 DOI: 10.1016/j.exphem.2019.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 11/18/2022]
Abstract
In contrast to steady-state erythropoiesis, which generates new erythrocytes at a constant rate, stress erythropoiesis rapidly produces a large bolus of new erythrocytes in response to anemic stress. In this study, we illustrate that Yes-associated protein (Yap1) promotes the rapid expansion of a transit-amplifying population of stress erythroid progenitors in vivo and in vitro. Yap1-mutated erythroid progenitors failed to proliferate in the spleen after transplantation into lethally irradiated recipient mice. Additionally, loss of Yap1 impaired the growth of actively proliferating erythroid progenitors in vitro. This role in proliferation is supported by gene expression profiles showing that transiently amplifying stress erythroid progenitors express high levels of genes associated with Yap1 activity and genes induced by Yap1. Furthermore, Yap1 promotes the proliferation of stress erythroid progenitors in part by regulating the expression of key glutamine-metabolizing enzymes. Thus, Yap1 acts as an erythroid regulator that coordinates the metabolic status with the proliferation of erythroid progenitors to promote stress erythropoiesis.
Collapse
Affiliation(s)
- Siyang Hao
- Graduate Program in Molecular, Cellular and Integrative Biosciences, Penn State University, University Park, PA; Center for Molecular Immunology and Infectious Disease at Penn State University, University Park, PA
| | - Yurika Matsui
- Graduate Program in Molecular, Cellular and Integrative Biosciences, Penn State University, University Park, PA
| | - Zhi-Chun Lai
- Graduate Program in Molecular, Cellular and Integrative Biosciences, Penn State University, University Park, PA; Graduate Program in Biochemistry, Microbiology and Molecular Biology, Penn State University, University Park, PA; Department of Biology, Penn State University, University Park, PA
| | - Robert F Paulson
- Graduate Program in Molecular, Cellular and Integrative Biosciences, Penn State University, University Park, PA; Center for Molecular Immunology and Infectious Disease at Penn State University, University Park, PA; Graduate Program in Biochemistry, Microbiology and Molecular Biology, Penn State University, University Park, PA; Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA.
| |
Collapse
|
21
|
Stoner SA, Yan M, Liu KTH, Arimoto KI, Shima T, Wang HY, Johnson DT, Bejar R, Jamieson C, Guan KL, Zhang DE. Hippo kinase loss contributes to del(20q) hematologic malignancies through chronic innate immune activation. Blood 2019; 134:1730-1744. [PMID: 31434702 PMCID: PMC6856986 DOI: 10.1182/blood.2019000170] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/09/2019] [Indexed: 12/13/2022] Open
Abstract
Heterozygous deletions within chromosome 20q, or del(20q), are frequent cytogenetic abnormalities detected in hematologic malignancies. To date, identification of genes in the del(20q) common deleted region that contribute to disease development have remained elusive. Through assessment of patient gene expression, we have identified STK4 (encoding Hippo kinase MST1) as a 20q gene that is downregulated below haploinsufficient amounts in myelodysplastic syndrome (MDS) and myeloproliferative neoplasm (MPN). Hematopoietic-specific gene inactivation in mice revealed Hippo kinase loss to induce splenomegaly, thrombocytopenia, megakaryocytic dysplasia, and a propensity for chronic granulocytosis; phenotypes that closely resemble those observed in patients harboring del(20q). In a JAK2-V617F model, heterozygous Hippo kinase inactivation led to accelerated development of lethal myelofibrosis, recapitulating adverse MPN disease progression and revealing a novel genetic interaction between these 2 molecular events. Quantitative serum protein profiling showed that myelofibrotic transformation in mice was associated with cooperative effects of JAK2-V617F and Hippo kinase inactivation on innate immune-associated proinflammatory cytokine production, including IL-1β and IL-6. Mechanistically, MST1 interacted with IRAK1, and shRNA-mediated knockdown was sufficient to increase IRAK1-dependent innate immune activation of NF-κB in human myeloid cells. Consistent with this, treatment with a small molecule IRAK1/4 inhibitor rescued the aberrantly elevated IL-1β production in the JAK2-V617F MPN model. This study identified Hippo kinase MST1 (STK4) as having a central role in the biology of del(20q)-associated hematologic malignancies and revealed a novel molecular basis of adverse MPN progression that may be therapeutically exploitable via IRAK1 inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rafael Bejar
- Moores Cancer Center
- Biomedical Sciences Graduate Program
- Division of Hematology and Oncology, Department of Medicine
| | - Catriona Jamieson
- Moores Cancer Center
- Biomedical Sciences Graduate Program
- Division of Regenerative Medicine, Department of Medicine, and
| | - Kun-Liang Guan
- Moores Cancer Center
- Biomedical Sciences Graduate Program
- Department of Pharmacology, University of California San Diego, La Jolla, CA
| | - Dong-Er Zhang
- Moores Cancer Center
- Biomedical Sciences Graduate Program
- Division of Biological Sciences
- Department of Pathology
| |
Collapse
|
22
|
Lodge EJ, Santambrogio A, Russell JP, Xekouki P, Jacques TS, Johnson RL, Thavaraj S, Bornstein SR, Andoniadou CL. Homeostatic and tumourigenic activity of SOX2+ pituitary stem cells is controlled by the LATS/YAP/TAZ cascade. eLife 2019; 8:43996. [PMID: 30912742 PMCID: PMC6461440 DOI: 10.7554/elife.43996] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/25/2019] [Indexed: 12/14/2022] Open
Abstract
SOX2 positive pituitary stem cells (PSCs) are specified embryonically and persist throughout life, giving rise to all pituitary endocrine lineages. We have previously shown the activation of the STK/LATS/YAP/TAZ signalling cascade in the developing and postnatal mammalian pituitary. Here, we investigate the function of this pathway during pituitary development and in the regulation of the SOX2 cell compartment. Through loss- and gain-of-function genetic approaches, we reveal that restricting YAP/TAZ activation during development is essential for normal organ size and specification from SOX2+ PSCs. Postnatal deletion of LATS kinases and subsequent upregulation of YAP/TAZ leads to uncontrolled clonal expansion of the SOX2+ PSCs and disruption of their differentiation, causing the formation of non-secreting, aggressive pituitary tumours. In contrast, sustained expression of YAP alone results in expansion of SOX2+ PSCs capable of differentiation and devoid of tumourigenic potential. Our findings identify the LATS/YAP/TAZ signalling cascade as an essential component of PSC regulation in normal pituitary physiology and tumourigenesis. The pituitary is a gland inside the head that releases hormones that control major processes in the body including growth, fertility and stress. Diseases of the pituitary gland can prevent the body from producing the appropriate amounts of hormones, and also include tumours. A population of stem cells in the pituitary known as SOX2 cells divide to make the specialist cells that produce the hormones. This population forms as the pituitary develops in the embryo and continues to contribute new hormone-producing cells throughout life. Signals from inside and outside the gland control how the pituitary develops and maintain the correct balance of different types of cells in the gland in adults. In 2016, Lodge et al. reported that a cascade of signals known as the Hippo pathway is active in mouse and human pituitary glands, but its role remained unclear. Here, Lodge et al. use genetic approaches to study this signalling pathway in the pituitary of mice. The results of the experiments show that the Hippo pathway is essential for the pituitary gland to develop normally in mouse embryos. Furthermore, in adult mice the Hippo pathway is required to maintain the population of SOX2 cells in the pituitary and to regulate their cell numbers. Increasing the level of Hippo signalling in mouse embryos and adult mice led to an expansion of SOX2 stem cells that could generate new specialist cell types, but a further increase generated aggressive tumours that originated from the uncontrolled growth of SOX2 cells. These findings are the first step to understanding how the Hippo pathway works in the pituitary, which may eventually lead to new treatments for tumours and other diseases that affect this gland. The next step towards such treatments will be to carry out further experiments that use drugs to control this pathway and alter the fate of pituitary cells in mice and other animals.
Collapse
Affiliation(s)
- Emily J Lodge
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom.,Division of Diabetes & Nutritional Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Alice Santambrogio
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom.,Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - John P Russell
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom
| | - Paraskevi Xekouki
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom.,Department of Endocrinology, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Thomas S Jacques
- UCL GOS Institute of Child Health and Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Randy L Johnson
- Department of Cancer Biology, The University of Texas, MD Anderson Cancer Center, Houston, United States
| | - Selvam Thavaraj
- Centre for Oral, Clinical and Translational Sciences, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom
| | - Stefan R Bornstein
- Division of Diabetes & Nutritional Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom.,Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Cynthia Lilian Andoniadou
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom.,Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
23
|
Mamidi A, Prawiro C, Seymour PA, de Lichtenberg KH, Jackson A, Serup P, Semb H. Mechanosignalling via integrins directs fate decisions of pancreatic progenitors. Nature 2018; 564:114-118. [DOI: 10.1038/s41586-018-0762-2] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 10/19/2018] [Indexed: 12/18/2022]
|
24
|
Wu R, Yang H, Wan J, Deng X, Chen L, Hao S, Ma L. Knockdown of the Hippo transducer YAP reduces proliferation and promotes apoptosis in the Jurkat leukemia cell. Mol Med Rep 2018; 18:5379-5388. [PMID: 30320399 PMCID: PMC6236312 DOI: 10.3892/mmr.2018.9556] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 09/10/2018] [Indexed: 12/26/2022] Open
Abstract
Leukemia and lymphoma are common hematological malignancies in children and young adults, which pose a tremendous threat to the survival of these young patients worldwide, despite availability of various effective treatments. The Hippo pathway is a novel-signaling pathway that regulates organ size, cell proliferation, apoptosis and tumorigenesis. The chief component of this pathway is the transducer yes-associated protein (YAP) which is over-expressed in numerous categories of tumors. However, little is known about the effect of YAP in hematological malignancies. In the present study, YAP expression was screened in several leukemia and lymphoma cell lines, and high YAP expression was demonstrated in Jurkat cells. To further unravel its effect on the biological behavior of Jurkat cells, lentivirus transduced short hairpin RNA (shRNA) technique was used to silence YAP. As expected, the YAP-specific shRNA dramatically inhibited YAP expression at the mRNA and protein levels. Reduced leukemia cell proliferation and increased cell apoptosis were demonstrated in YAP knockdown Jurkat cells. It was also demonstrated that YAP knockdown resulted in deregulated expression of a cluster of downstream genes crucial to cell proliferation or apoptosis, including protein kinase B, B-cell lymphoma 2 (BCL2) and BCL2 like protein 1. Consequently, the results of the present study established that suppression of YAP expression serves an important role in Jurkat cell proliferation and apoptosis, which may serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Ran Wu
- Department of Hematology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Hui Yang
- Department of Hematology, Shanghai 6th People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Jiangbo Wan
- Department of Hematology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Xiaohui Deng
- Department of Hematology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Linjun Chen
- Department of Hematology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Siguo Hao
- Department of Hematology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Liyuan Ma
- Department of Hematology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
25
|
Petersen N, Frimurer TM, Terndrup Pedersen M, Egerod KL, Wewer Albrechtsen NJ, Holst JJ, Grapin-Botton A, Jensen KB, Schwartz TW. Inhibiting RHOA Signaling in Mice Increases Glucose Tolerance and Numbers of Enteroendocrine and Other Secretory Cells in the Intestine. Gastroenterology 2018; 155:1164-1176.e2. [PMID: 29935151 DOI: 10.1053/j.gastro.2018.06.039] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Glucagon-like peptide 1 (GLP1) is produced by L cells in the intestine, and agonists of the GLP1 receptor are effective in the treatment of diabetes. Levels of GLP1 increase with numbers of L cells. Therefore, agents that increase numbers of L cell might be developed for treatment of diabetes. Ras homologue family member A (RhoA) signaling through Rho-associated coiled-coil-containing protein kinases 1 and 2 (ROCK1 and ROCK2) controls cell differentiation, but it is not clear whether this pathway regulates enteroendocrine differentiation in the intestinal epithelium. We investigated the effects of Y-27632, an inhibitor of ROCK1 and ROCK2, on L-cell differentiation. METHODS We collected intestinal tissues from GLU-Venus, GPR41-RFP, and Neurog3-RFP mice, in which the endocrine lineage is fluorescently labeled, for in vitro culture and histologic analysis. Small intestine organoids derived from these mice were cultured with Y-27632 and we measured percentages of L cells, expression of intestinal cell-specific markers, and secretion of GLP1 in medium. Mice were fed a normal chow or a high-fat diet and given Y-27632 or saline (control) and blood samples were collected for measurement of GLP1, insulin, and glucose. RESULTS Incubation of intestinal organoids with Y-27632 increased numbers of L cells and secretion of GLP1. These increases were associated with upregulated expression of genes encoding intestinal hormones, neurogenin 3, neurogenic differentiation factor 1, forkhead box A1 and A2, and additional markers of secretory cells. Mice fed the normal chow diet and given Y-27632 had increased numbers of L cells in intestinal tissues, increased plasma levels of GLP1 and insulin, and lower blood levels of glucose compared with mice fed the normal chow diet and given saline. In mice with insulin resistance induced by the high-fat diet, administration of Y-27632 increased secretion of GLP1 and glucose tolerance compared with administration of saline. CONCLUSIONS In mouse intestinal organoids, an inhibitor of RhoA signaling increased the differentiation of the secretory lineage and the development of enteroendocrine cells. Inhibitors of RhoA signaling or other strategies to increase numbers of L cells might be developed for treatment of patients with type 2 diabetes or for increasing glucose tolerance.
Collapse
Affiliation(s)
- Natalia Petersen
- Section of Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Thomas M Frimurer
- Section of Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Kristoffer L Egerod
- Section of Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences and the Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Translational Metabolic Physiology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences and the Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Translational Metabolic Physiology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Grapin-Botton
- Novo Nordisk Foundation Center for Stem Cell Research, Faculty of Medical and Health, University of Copenhagen, Copenhagen, Denmark
| | - Kim B Jensen
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Stem Cell Research, Faculty of Medical and Health, University of Copenhagen, Copenhagen, Denmark
| | - Thue W Schwartz
- Section of Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
A Computational Method for Classifying Different Human Tissues with Quantitatively Tissue-Specific Expressed Genes. Genes (Basel) 2018; 9:genes9090449. [PMID: 30205473 PMCID: PMC6162521 DOI: 10.3390/genes9090449] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/01/2018] [Accepted: 09/04/2018] [Indexed: 02/06/2023] Open
Abstract
Tissue-specific gene expression has long been recognized as a crucial key for understanding tissue development and function. Efforts have been made in the past decade to identify tissue-specific expression profiles, such as the Human Proteome Atlas and FANTOM5. However, these studies mainly focused on "qualitatively tissue-specific expressed genes" which are highly enriched in one or a group of tissues but paid less attention to "quantitatively tissue-specific expressed genes", which are expressed in all or most tissues but with differential expression levels. In this study, we applied machine learning algorithms to build a computational method for identifying "quantitatively tissue-specific expressed genes" capable of distinguishing 25 human tissues from their expression patterns. Our results uncovered the expression of 432 genes as optimal features for tissue classification, which were obtained with a Matthews Correlation Coefficient (MCC) of more than 0.99 yielded by a support vector machine (SVM). This constructed model was superior to the SVM model using tissue enriched genes and yielded MCC of 0.985 on an independent test dataset, indicating its good generalization ability. These 432 genes were proven to be widely expressed in multiple tissues and a literature review of the top 23 genes found that most of them support their discriminating powers. As a complement to previous studies, our discovery of these quantitatively tissue-specific genes provides insights into the detailed understanding of tissue development and function.
Collapse
|
27
|
Zhu JY, Lin S, Ye J. YAP and TAZ, the conductors that orchestrate eye development, homeostasis, and disease. J Cell Physiol 2018; 234:246-258. [PMID: 30094836 DOI: 10.1002/jcp.26870] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/08/2018] [Accepted: 05/18/2018] [Indexed: 12/25/2022]
Abstract
Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are transcriptional coactivators established as a nexus in numerous signaling pathways, notably in Hippo signaling. Previous research revealed multifarious function of YAP and TAZ in oncology and cardiovasology. Recently, the focus has been laid on their pivotal role in eye morphogenesis and homeostasis. In this review, we synthesize advances of YAP and TAZ function during eye development in different model organisms, introduce their function in different ocular tissues and eye diseases, and highlight the potential for therapeutic interventions.
Collapse
Affiliation(s)
- Jing-Yi Zhu
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Sen Lin
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Jian Ye
- Department of Ophthalmology and Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
28
|
Li H, Chen C, Wang D. Low‑frequency ultrasound and microbubbles combined with simvastatin promote the apoptosis of MCF‑7 cells by affecting the LATS1/YAP/RHAMM pathway. Mol Med Rep 2018; 18:2724-2732. [PMID: 30015955 PMCID: PMC6102727 DOI: 10.3892/mmr.2018.9273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 12/13/2017] [Indexed: 12/17/2022] Open
Abstract
Ultrasound scanning has widespread used in clinical practice and also has therapeutic applications. Simvastatin is a statins that is able to competitively inhibit the activity of 3‑hydroxy‑3‑methylglutaryl‑coenzyme A reductase. The aim of the present study was to investigate the roles and mechanisms of low‑frequency ultrasound (LFU) and microbubbles combined with simvastatin on MCF‑7 cell growth and apoptosis. Cell viability, apoptosis and cell cycle were evaluated using an MTT assay and flow cytometry, respectively. The expression of related proteins was measured by western blot assay. The results revealed that simvastatin and LFU with microbubbles reduces the viability of MCF‑7 cells. The combination of LFU and microbubbles with simvastatin promoted the apoptosis of MCF‑7 cells. Furthermore, it was confirmed that LFU and microbubbles combined with simvastatin affected the large tumor suppressor 1 (LATS1)/yes‑associated protein (YAP)/receptor of the hyaluronan‑mediated motility (RHAMM) pathway in MCF‑7 cells. It was determined that LATS1 acts as a negative regulator in the LATS1/YAP/RHAMM pathway in MCF‑7 cells. In conclusion, the results of the present study indicate that LFU and microbubbles combined with simvastatin promotes the apoptosis of MCF‑7 cells via the LATS1/YAP/RHAMM pathway. The present study suggested a possible strategy for the treatment of breast cancer.
Collapse
Affiliation(s)
- Haige Li
- Department of Imaging, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Chen Chen
- Department of Imaging, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Dehang Wang
- Department of Imaging, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
29
|
Moon S, Yeon Park S, Woo Park H. Regulation of the Hippo pathway in cancer biology. Cell Mol Life Sci 2018; 75:2303-2319. [PMID: 29602952 PMCID: PMC11105795 DOI: 10.1007/s00018-018-2804-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/17/2018] [Accepted: 03/22/2018] [Indexed: 01/23/2023]
Abstract
The Hippo tumor suppressor pathway, which is well conserved from Drosophila to humans, has emerged as the master regulator of organ size, as well as major cellular properties, such as cell proliferation, survival, stemness, and tissue homeostasis. The biological significance and deregulation of the Hippo pathway in tumorigenesis have received a surge of interest in the past decade. In the current review, we present the major discoveries that made substantial contributions to our understanding of the Hippo pathway and discuss how Hippo pathway components contribute to cellular signaling, physiology, and their potential implications in anticancer therapeutics.
Collapse
Affiliation(s)
- Sungho Moon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - So Yeon Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
30
|
Li N, Tong X, Zeng J, Meng G, Sun F, Hu H, Song J, Lu C, Dai F. Hippo pathway regulates somatic development and cell proliferation of silkworm. Genomics 2018; 111:391-397. [PMID: 29501778 DOI: 10.1016/j.ygeno.2018.02.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/06/2018] [Accepted: 02/23/2018] [Indexed: 01/20/2023]
Abstract
Hippo signaling pathway (signaling pathway Hippo, hereinafter referred to as the Hippo pathway) was the earliest found in Drosophila (Schneck [1]), which can regulate the development of tissues and organs, even some components of the pathway were identified as tumor suppressor [2]. The pathway was more concerned in fruit flies and mice (Schneck [1]), but little attention has been given in silkworm, an important economic and lepidopteran model insect. In this manuscript, we identified major Hippo pathway related genes (Hippo, Salvador, Warts, Mats, Yorkie) in silkworm and named BmHpo, BmSav, BmWts, BmMats, BmYki. The domain organization of these genes was highly conserved in silkworm and other organisms suggesting that they could use similar protein-protein interactions to construct the Hippo kinase cascades. The expression profiles of these genes in silkworm during embryonic, larval, wandering, pupal and adult stages were analyzed, 14 tissues/organs of the day 3, 5th instar larvae (L5D3) as well. Experimental results showed that the expression of Hippo pathway had some influence on the development of silkworm. In order to find out the mechanism of Hippo pathway affecting silkworm development, BmHpo and BmYki were up-regulated and de-regulated in the cell line of Bombyx mori-BmN-SWU1(NS), and the changes of cell proliferation activity and cell cycle were detected. The distribution of BmYki was detected by immunofluorescence technique. This study provides insights into the genes of Hippo pathway which have a certain effect on somatic development and cell proliferation in silkworm.
Collapse
Affiliation(s)
- Niannian Li
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400715, China
| | - Xiaoling Tong
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400715, China
| | - Jie Zeng
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400715, China
| | - Gang Meng
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400715, China
| | - Fuze Sun
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400715, China
| | - Hai Hu
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400715, China
| | - Jiangbo Song
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400715, China
| | - Cheng Lu
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400715, China
| | - Fangyin Dai
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing 400715, China.
| |
Collapse
|
31
|
Sharif AA, Hergovich A. The NDR/LATS protein kinases in immunology and cancer biology. Semin Cancer Biol 2018; 48:104-114. [PMID: 28579171 DOI: 10.1016/j.semcancer.2017.04.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/15/2017] [Accepted: 04/25/2017] [Indexed: 02/07/2023]
|
32
|
Croci O, De Fazio S, Biagioni F, Donato E, Caganova M, Curti L, Doni M, Sberna S, Aldeghi D, Biancotto C, Verrecchia A, Olivero D, Amati B, Campaner S. Transcriptional integration of mitogenic and mechanical signals by Myc and YAP. Genes Dev 2017; 31:2017-2022. [PMID: 29141911 PMCID: PMC5733494 DOI: 10.1101/gad.301184.117] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [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: 12/12/2022]
Abstract
The transcription factors Myc and YAP–TEAD act downstream from mitogenic signals, with the latter responding also to mechanical cues. Here, Croci et al. show that these factors coordinately regulate genes required for cell proliferation. Mammalian cells must integrate environmental cues to determine coherent physiological responses. The transcription factors Myc and YAP–TEAD act downstream from mitogenic signals, with the latter responding also to mechanical cues. Here, we show that these factors coordinately regulate genes required for cell proliferation. Activation of Myc led to extensive association with its genomic targets, most of which were prebound by TEAD. At these loci, recruitment of YAP was Myc-dependent and led to full transcriptional activation. This cooperation was critical for cell cycle entry, organ growth, and tumorigenesis. Thus, Myc and YAP–TEAD integrate mitogenic and mechanical cues at the transcriptional level to provide multifactorial control of cell proliferation.
Collapse
Affiliation(s)
- Ottavio Croci
- Center for Genomic Science of IIT@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Serena De Fazio
- Center for Genomic Science of IIT@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Francesca Biagioni
- Center for Genomic Science of IIT@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Elisa Donato
- Center for Genomic Science of IIT@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Marieta Caganova
- Center for Genomic Science of IIT@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Laura Curti
- Center for Genomic Science of IIT@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Mirko Doni
- Department of Experimental Oncology, European Institute of Oncology (IEO), 20139 Milan, Italy
| | - Silvia Sberna
- Center for Genomic Science of IIT@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Deborah Aldeghi
- Center for Genomic Science of IIT@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Chiara Biancotto
- Center for Genomic Science of IIT@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Alessandro Verrecchia
- Department of Experimental Oncology, European Institute of Oncology (IEO), 20139 Milan, Italy
| | - Daniela Olivero
- Laboratorio di Analisi Veterinarie BiEsseA, 20129 Milan, Italy
| | - Bruno Amati
- Center for Genomic Science of IIT@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy.,Department of Experimental Oncology, European Institute of Oncology (IEO), 20139 Milan, Italy
| | - Stefano Campaner
- Center for Genomic Science of IIT@SEMM (Istituto Italiano di Tecnologia at European School of Molecular Medicine), Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| |
Collapse
|
33
|
Redondo PA, Pavlou M, Loizidou M, Cheema U. Elements of the niche for adult stem cell expansion. J Tissue Eng 2017; 8:2041731417725464. [PMID: 28890779 PMCID: PMC5574483 DOI: 10.1177/2041731417725464] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 07/18/2017] [Indexed: 12/21/2022] Open
Abstract
Adult stem cells are crucial for tissue homeostasis. These cells reside within exclusive locations in tissues, termed niches, which protect adult stem cell fidelity and regulate their many functions through biophysical-, biochemical- and cellular-mediated mechanisms. There is a growing understanding of how these mechanisms and their components contribute towards maintaining stem cell quiescence, self-renewal, expansion and differentiation patterns. In vitro expansion of adult stem cells is a powerful tool for understanding stem cell biology, and for tissue engineering and regenerative medicine applications. However, it is technically challenging, since adult stem cell removal from their native microenvironment has negative repercussions on their sustainability. In this review, we overview specific elements of the biomimetic niche and how recreating such elements can help in vitro propagation of adult stem cells.
Collapse
Affiliation(s)
- Patricia A Redondo
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Marina Pavlou
- Institute of Orthopaedics & Musculoskeletal Science, University College London, London, UK
| | - Marilena Loizidou
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Umber Cheema
- Institute of Orthopaedics & Musculoskeletal Science, University College London, London, UK
| |
Collapse
|
34
|
Ahmed AA, Mohamed AD, Gener M, Li W, Taboada E. YAP and the Hippo pathway in pediatric cancer. Mol Cell Oncol 2017; 4:e1295127. [PMID: 28616573 DOI: 10.1080/23723556.2017.1295127] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/01/2017] [Accepted: 02/10/2017] [Indexed: 12/17/2022]
Abstract
The Hippo pathway is an important signaling pathway that controls cell proliferation and apoptosis. It is evolutionarily conserved in mammals and is stimulated by cell-cell contact, inhibiting cell proliferation in response to increased cell density. During early embryonic development, the Hippo signaling pathway regulates organ development and size, and its functions result in the coordinated balance between proliferation, apoptosis, and differentiation. Its principal effectors, YAP and TAZ, regulate signaling by the embryonic stem cells and determine cell fate and histogenesis. Dysfunction of this pathway contributes to cancer development in adults and children. Emerging studies have shed light on the upregulation of Hippo pathway members in several pediatric cancers and may offer prognostic information on rhabdomyosarcoma, osteosarcoma, Wilms tumor, neuroblastoma, medulloblastoma, and other brain gliomas. We review the results of such published studies and highlight the potential clinical application of this pathway in pediatric oncologic and pathologic studies. These studies support targeting this pathway as a novel treatment strategy.
Collapse
Affiliation(s)
- Atif A Ahmed
- Department of Pathology, Children's Mercy Hospital, Kansas City, MO, USA
| | | | - Melissa Gener
- Department of Pathology, Children's Mercy Hospital, Kansas City, MO, USA
| | - Weijie Li
- Department of Pathology, Children's Mercy Hospital, Kansas City, MO, USA
| | - Eugenio Taboada
- Department of Pathology, Children's Mercy Hospital, Kansas City, MO, USA
| |
Collapse
|
35
|
Nishio M, Maehama T, Goto H, Nakatani K, Kato W, Omori H, Miyachi Y, Togashi H, Shimono Y, Suzuki A. Hippo vs. Crab: tissue-specific functions of the mammalian Hippo pathway. Genes Cells 2017; 22:6-31. [PMID: 28078823 DOI: 10.1111/gtc.12461] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 11/18/2016] [Indexed: 12/13/2022]
Abstract
The Hippo signaling pathway is a vital suppressor of tumorigenesis that is often inactivated in human cancers. In normal cells, the Hippo pathway is triggered by external forces such as cell crowding, or changes to the extracellular matrix or cell polarity. Once activated, Hippo signaling down-regulates transcription supported by the paralogous cofactors YAP1 and TAZ. The Hippo pathway's functions in normal and cancer biology have been dissected by studies of mutant mice with null or conditional tissue-specific mutations of Hippo signaling elements. In this review, we attempt to systematically summarize results that have been gleaned from detailed in vivo characterizations of these mutants. Our goal is to describe the physiological roles of Hippo signaling in several normal organ systems, as well as to emphasize how disruption of the Hippo pathway, and particularly hyperactivation of YAP1/TAZ, can be oncogenic.
Collapse
Affiliation(s)
- Miki Nishio
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Japan.,Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Tomohiko Maehama
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroki Goto
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Keisuke Nakatani
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Wakako Kato
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hirofumi Omori
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yosuke Miyachi
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hideru Togashi
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yohei Shimono
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akira Suzuki
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Japan.,Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
36
|
Passaniti A, Brusgard JL, Qiao Y, Sudol M, Finch-Edmondson M. Roles of RUNX in Hippo Pathway Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 962:435-448. [PMID: 28299672 DOI: 10.1007/978-981-10-3233-2_26] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The Runt-domain (RD) transcription factors (RUNX genes) are an important family of transcriptional mediators that interact with a variety of proteins including the Hippo pathway effector proteins, YAP and TAZ. In this chapter we focus on two examples of RUNX-TAZ/YAP interactions that have particular significance in human cancer. Specifically, recent evidence has found that RUNX2 cooperates with TAZ to promote epithelial to mesenchymal transition mediated by the soluble N-terminal ectodomain of E-Cadherin, sE-Cad. Contrastingly, in gastric cancer, RUNX3 acts as a tumor suppressor via inhibition of the YAP-TEAD complex and disruption of downstream YAP-mediated gene transcription and the oncogenic phenotype. The reports highlighted in this chapter add to the growing repertoire of instances of Hippo pathway crosstalk that have been identified in cancer. Elucidation of these increasingly complex interactions may help to identify novel strategies to target Hippo pathway dysregulation in human cancer.
Collapse
Affiliation(s)
- Antonino Passaniti
- Department of Pathology and Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, and the Veterans Administration Health Service, Baltimore, MD, USA.
| | - Jessica L Brusgard
- Department of Pathology and Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, and the Veterans Administration Health Service, Baltimore, MD, USA
| | - Yiting Qiao
- The Mechanobiology Institute (MBI) and the NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Marius Sudol
- The Mechanobiology Institute (MBI) and the NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Institute of Molecular and Cell Biology A*STAR, Singapore, Republic of Singapore
| | - Megan Finch-Edmondson
- The Mechanobiology Institute (MBI) and the NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| |
Collapse
|
37
|
Cacemiro MDC, Berzoti-Coelho MG, Cominal JG, Burin SM, Castro FAD. Hippo pathway deregulation: implications in the pathogenesis of haematological malignancies. J Clin Pathol 2016; 70:9-14. [PMID: 27798082 DOI: 10.1136/jclinpath-2016-204055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/26/2016] [Accepted: 09/27/2016] [Indexed: 11/04/2022]
Abstract
The Hippo pathway participates in the regulation of cell proliferation, differentiation and apoptosis. It is composed by a large array of proteins whose deregulation has been associated with pro-oncogenic and antioncogenic processes. The present review focuses on the Hippo pathway signalling network and discusses its dual role in oncogenesis, particularly in haematological malignancies.
Collapse
Affiliation(s)
- Maira da Costa Cacemiro
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, São Paulo, Brazil
| | - Maria Gabriela Berzoti-Coelho
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, São Paulo, Brazil
| | - Juçara Gastaldi Cominal
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, São Paulo, Brazil
| | - Sandra Mara Burin
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, São Paulo, Brazil
| | - Fabíola Attié de Castro
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
38
|
Qiu J, Zhou B, Thol F, Zhou Y, Chen L, Shao C, DeBoever C, Hou J, Li H, Chaturvedi A, Ganser A, Bejar R, Zhang DE, Fu XD, Heuser M. Distinct splicing signatures affect converged pathways in myelodysplastic syndrome patients carrying mutations in different splicing regulators. RNA (NEW YORK, N.Y.) 2016; 22:1535-1549. [PMID: 27492256 PMCID: PMC5029452 DOI: 10.1261/rna.056101.116] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 06/08/2016] [Indexed: 06/06/2023]
Abstract
Myelodysplastic syndromes (MDS) are heterogeneous myeloid disorders with prevalent mutations in several splicing factors, but the splicing programs linked to specific mutations or MDS in general remain to be systematically defined. We applied RASL-seq, a sensitive and cost-effective platform, to interrogate 5502 annotated splicing events in 169 samples from MDS patients or healthy individuals. We found that splicing signatures associated with normal hematopoietic lineages are largely related to cell signaling and differentiation programs, whereas MDS-linked signatures are primarily involved in cell cycle control and DNA damage responses. Despite the shared roles of affected splicing factors in the 3' splice site definition, mutations in U2AF1, SRSF2, and SF3B1 affect divergent splicing programs, and interestingly, the affected genes fall into converging cancer-related pathways. A risk score derived from 11 splicing events appears to be independently associated with an MDS prognosis and AML transformation, suggesting potential clinical relevance of altered splicing patterns in MDS.
Collapse
Affiliation(s)
- Jinsong Qiu
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Bing Zhou
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Felicitas Thol
- Department of Hematology, Hemostasis, Oncology and Stem cell Transplantation, Hannover Medical School, 30625 Hannover, Germany
| | - Yu Zhou
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Liang Chen
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Changwei Shao
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Christopher DeBoever
- Institute for Genomic Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Jiayi Hou
- Clinical and Translational Research Institute, University of California, San Diego, La Jolla, California 92093, USA
| | - Hairi Li
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Anuhar Chaturvedi
- Department of Hematology, Hemostasis, Oncology and Stem cell Transplantation, Hannover Medical School, 30625 Hannover, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology and Stem cell Transplantation, Hannover Medical School, 30625 Hannover, Germany
| | - Rafael Bejar
- Division of Hematology-Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Dong-Er Zhang
- Department of Pathology, Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Xiang-Dong Fu
- Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA Institute for Genomic Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem cell Transplantation, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
39
|
Cabochette P, Vega-Lopez G, Bitard J, Parain K, Chemouny R, Masson C, Borday C, Hedderich M, Henningfeld KA, Locker M, Bronchain O, Perron M. YAP controls retinal stem cell DNA replication timing and genomic stability. eLife 2015; 4:e08488. [PMID: 26393999 PMCID: PMC4578106 DOI: 10.7554/elife.08488] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 08/20/2015] [Indexed: 12/27/2022] Open
Abstract
The adult frog retina retains a reservoir of active neural stem cells that contribute to continuous eye growth throughout life. We found that Yap, a downstream effector of the Hippo pathway, is specifically expressed in these stem cells. Yap knock-down leads to an accelerated S-phase and an abnormal progression of DNA replication, a phenotype likely mediated by upregulation of c-Myc. This is associated with an increased occurrence of DNA damage and eventually p53-p21 pathway-mediated cell death. Finally, we identified PKNOX1, a transcription factor involved in the maintenance of genomic stability, as a functional and physical interactant of YAP. Altogether, we propose that YAP is required in adult retinal stem cells to regulate the temporal firing of replication origins and quality control of replicated DNA. Our data reinforce the view that specific mechanisms dedicated to S-phase control are at work in stem cells to protect them from genomic instability. DOI:http://dx.doi.org/10.7554/eLife.08488.001 In animals, stem cells divide to produce the new cells needed to grow and renew tissues and organs. Understanding the biology of these cells is of the utmost importance for developing new treatments for a wide range of human diseases, including neurodegenerative diseases and cancer. Before a stem cell divides, it copies its DNA and the two sets of genetic instructions are then separated so that the two daughter cells both have a complete set. This process needs to be as accurate as possible because any errors would result in incorrect genetic information being passed on to the daughter cells. Stem cells in the light-sensitive part of the eye—called the retina—divide to produce the cells that detect light and relay visual information to the brain. In many animals, these stem cells stop dividing soon after birth and the retina stops growing. However, the stem cells in frogs and fish continue to divide throughout the life of the animal, which enables the eye to keep on growing. A protein called YAP regulates the growth of organs in animal embryos, but it is not clear what role this protein plays in stem cells, particularly after birth. To address this question, Cabochette et al. studied YAP in the retina of frog tadpoles. The experiments show that YAP is produced in the stem cells of the retina after birth and is required for the retina to continue to grow. Cabochette et al. used tools called ‘photo-cleavable Morpholinos’ to alter the production of YAP in adult stem cells. The cells that produced less YAP copied their DNA more quickly and more of their DNA became damaged, which eventually led to the death of these cells. Further experiments revealed that YAP interacts with a protein called PKNOX1, which is involved in maintaining the integrity of DNA. Cabochette et al.'s findings provide the first insights into how YAP works in the stem cells of the retina and demonstrate that it plays a crucial role in regulating when DNA is copied. A future challenge is to find out whether YAP plays a similar role in the stem cells of other organs in adult animals. DOI:http://dx.doi.org/10.7554/eLife.08488.002
Collapse
Affiliation(s)
- Pauline Cabochette
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Orsay, France
| | - Guillermo Vega-Lopez
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Orsay, France
| | - Juliette Bitard
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Orsay, France
| | - Karine Parain
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Institute of Developmental Biochemistry, University of Goettingen, Goettingen, Germany
| | - Romain Chemouny
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Orsay, France
| | - Christel Masson
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Orsay, France
| | - Caroline Borday
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Orsay, France
| | - Marie Hedderich
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Institute of Developmental Biochemistry, University of Goettingen, Goettingen, Germany
| | - Kristine A Henningfeld
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Institute of Developmental Biochemistry, University of Goettingen, Goettingen, Germany
| | - Morgane Locker
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Orsay, France
| | - Odile Bronchain
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Orsay, France
| | - Muriel Perron
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Orsay, France
| |
Collapse
|
40
|
Shin JW, Discher DE. Blood and immune cell engineering: Cytoskeletal contractility and nuclear rheology impact cell lineage and localization: Biophysical regulation of hematopoietic differentiation and trafficking. Bioessays 2015; 37:633-42. [PMID: 25810145 DOI: 10.1002/bies.201400166] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Clinical success with human hematopoietic stem cell (HSC) transplantation establishes a paradigm for regenerative therapies with other types of stem cells. However, it remains generally challenging to therapeutically treat tissues after engineering of stem cells in vitro. Recent studies suggest that stem and progenitor cells sense physical features of their niches. Here, we review biophysical contributions to lineage decisions, maturation, and trafficking of blood and immune cells. Polarized cellular contractility and nuclear rheology are separately shown to be functional markers of a hematopoietic hierarchy that predict the ability of a lineage to traffic in and out of the bone marrow niche. These biophysical determinants are regulated by a set of structural molecules, including cytoplasmic myosin-II and nuclear lamins, which themselves are modulated by a diverse range of transcriptional and post-translational mechanisms. Small molecules that target these mechanobiological circuits, along with novel bioengineering methods, could prove broadly useful in programming blood and immune cells for therapies ranging from blood transfusions to immune attack of tumors.
Collapse
Affiliation(s)
- Jae-Won Shin
- Biophysical Engineering Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| | - Dennis E Discher
- Biophysical Engineering Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
41
|
Cheng Y, Feng Y, Jansson L, Sato Y, Deguchi M, Kawamura K, Hsueh AJ. Actin polymerization-enhancing drugs promote ovarian follicle growth mediated by the Hippo signaling effector YAP. FASEB J 2015; 29:2423-30. [PMID: 25690654 DOI: 10.1096/fj.14-267856] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/27/2015] [Indexed: 11/11/2022]
Abstract
Hippo signaling pathway consists of conserved serine/threonine kinases to maintain optimal organ sizes. Studies have demonstrated that fragmentation of murine ovaries increases actin polymerization and disrupts Hippo signaling, leading to nuclear translocation of Hippo signaling effector Yes-associated protein (YAP) in ovarian follicles and follicle growth. For patients with polycystic ovarian syndrome showing follicle arrest, ovarian wedge resection and laser drilling promote follicle growth. Because these damaging procedures likely involve actin polymerization, we tested whether actin polymerization-promoting drugs could promote YAP translocation and stimulate follicle growth. Treatment of murine ovaries with μM Jasplakinolide (JASP), an actin polymerization-promoting cyclic peptide, or sphingosine-1-phosphate (S1P), a follicular fluid constituent known to promote actin polymerization, increased the conversion of globular actin to the filamentous form, followed by increased nuclear YAP and expression of downstream connective tissue growth factor (CCN2). After short-term treatments with JASP or S1P, in vitro cultured and in vivo grafted ovaries showed follicle growth. Furthermore, induction of constitutively active YAP in ovarian grafts of transgenic mice enhanced follicle development, whereas treatment of human ovarian cortices with JASP or S1P increased CCN2 expression. Thus, JASP and S1P stimulate follicle growth and are potential therapeutic agents for treating polycystic ovarian syndrome and other ovarian disorders.
Collapse
Affiliation(s)
- Yuan Cheng
- *Department of Obstetrics and Gynecology and Department of Otolaryngology, Stanford University School of Medicine, Stanford, California, USA; Department of Obstetrics and Gynecology, St. Mariana University, Kawasaki, Japan; and Department of Obstetrics and Gynecology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yi Feng
- *Department of Obstetrics and Gynecology and Department of Otolaryngology, Stanford University School of Medicine, Stanford, California, USA; Department of Obstetrics and Gynecology, St. Mariana University, Kawasaki, Japan; and Department of Obstetrics and Gynecology, Akita University Graduate School of Medicine, Akita, Japan
| | - Lina Jansson
- *Department of Obstetrics and Gynecology and Department of Otolaryngology, Stanford University School of Medicine, Stanford, California, USA; Department of Obstetrics and Gynecology, St. Mariana University, Kawasaki, Japan; and Department of Obstetrics and Gynecology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yorino Sato
- *Department of Obstetrics and Gynecology and Department of Otolaryngology, Stanford University School of Medicine, Stanford, California, USA; Department of Obstetrics and Gynecology, St. Mariana University, Kawasaki, Japan; and Department of Obstetrics and Gynecology, Akita University Graduate School of Medicine, Akita, Japan
| | - Masashi Deguchi
- *Department of Obstetrics and Gynecology and Department of Otolaryngology, Stanford University School of Medicine, Stanford, California, USA; Department of Obstetrics and Gynecology, St. Mariana University, Kawasaki, Japan; and Department of Obstetrics and Gynecology, Akita University Graduate School of Medicine, Akita, Japan
| | - Kazuhiro Kawamura
- *Department of Obstetrics and Gynecology and Department of Otolaryngology, Stanford University School of Medicine, Stanford, California, USA; Department of Obstetrics and Gynecology, St. Mariana University, Kawasaki, Japan; and Department of Obstetrics and Gynecology, Akita University Graduate School of Medicine, Akita, Japan
| | - Aaron J Hsueh
- *Department of Obstetrics and Gynecology and Department of Otolaryngology, Stanford University School of Medicine, Stanford, California, USA; Department of Obstetrics and Gynecology, St. Mariana University, Kawasaki, Japan; and Department of Obstetrics and Gynecology, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
42
|
Affiliation(s)
- Fa-Xing Yu
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093;
- Children's Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China 200032
| | - Zhipeng Meng
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093;
| | - Steven W. Plouffe
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093;
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093;
| |
Collapse
|
43
|
Ferguson GB, Martinez-Agosto JA. Yorkie and Scalloped signaling regulates Notch-dependent lineage specification during Drosophila hematopoiesis. Curr Biol 2014; 24:2665-72. [PMID: 25454586 DOI: 10.1016/j.cub.2014.09.081] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 07/25/2014] [Accepted: 09/01/2014] [Indexed: 01/02/2023]
Abstract
Cellular microenvironments established by the spatial and temporal expression of specific signaling molecules are critical for both the maintenance and lineage-specific differentiation of progenitor cells. In Drosophila, a population of hematopoietic progenitors, or prohemocytes, within the larval lymph gland gives rise to three mature cell types: plasmatocytes, lamellocytes, and crystal cells. Removal of the secreted signaling molecules Hedgehog and PVF1 from the posterior signaling center (PSC), which acts as a niche, leads to a loss of progenitors and complete differentiation of the lymph gland. Here, we characterize a novel population of signaling cells within the lymph gland, distinct from the PSC, that are required for lineage-specific differentiation of crystal cells. We provide evidence that Yorkie and Scalloped, the Drosophila homologs of YAP and TEAD, are required in lineage-specifying cells to regulate expression of Serrate, the Notch ligand responsible for the initiation of the crystal cell differentiation program. Genetic manipulation of yorkie and scalloped in the lymph gland specifically alters Serrate expression and crystal cell differentiation. Furthermore, Serrate expression in lineage-specifying cells is eliminated in the lymph gland upon the immune response induced by wasp parasitization to ensure the proper differentiation of lamellocytes at the expense of crystal cells. These findings expand the roles for Yorkie/Scalloped beyond growth to encompass specific cell-fate determination in the context of blood development. Similar regulatory functions may extend to their homologs in vertebrate progenitor cell niches that are required for specifying cell fate.
Collapse
Affiliation(s)
- Gabriel B Ferguson
- Molecular Biology Interdepartmental Ph.D. Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Julian A Martinez-Agosto
- Molecular Biology Interdepartmental Ph.D. Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, Jonsson Comprehensive Cancer Center, UCLA Broad Stem Cell Center, and Mattel Children's Hospital UCLA, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
44
|
Abstract
Blood is renewed throughout the entire life. The stem cells of the blood, called hematopoietic stem cells (HSCs), are responsible for maintaining a supply of all types of fresh blood cells. In contrast to other stem cells, the clinical application of these cells is well established and HSC transplantation is an established life-saving therapy for patients suffering from haematological disorders. Despite their efficient functionality throughout life in vivo, controlling HSC behaviour in vitro (including their proliferation and differentiation) is still a major task that has not been resolved with standard cell culture systems. Targeted HSC multiplication in vitro could be beneficial for many patients, because HSC supply is limited. The biology of these cells and their natural microenvironment - their niche - remain a matter of ongoing research. In recent years, evidence has come to light that HSCs are susceptible to physical stimuli. This makes the regulation of HSCs by engineering physical parameters a promising approach for the targeted manipulation of these cells for clinical applications. Nevertheless, the biophysical regulation of these cells is still poorly understood. This review sheds light on the role of biophysical parameters in HSC biology and outlines which knowledge on biophysical regulation identified in other cell types could be applied to HSCs.
Collapse
Affiliation(s)
- C Lee-Thedieck
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany.
| | | |
Collapse
|
45
|
Huang J, Kalderon D. Coupling of Hedgehog and Hippo pathways promotes stem cell maintenance by stimulating proliferation. ACTA ACUST UNITED AC 2014; 205:325-38. [PMID: 24798736 PMCID: PMC4018789 DOI: 10.1083/jcb.201309141] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
It is essential to define the mechanisms by which external signals regulate adult stem cell numbers, stem cell maintenance, and stem cell proliferation to guide regenerative stem cell therapies and to understand better how cancers originate in stem cells. In this paper, we show that Hedgehog (Hh) signaling in Drosophila melanogaster ovarian follicle stem cells (FSCs) induces the activity of Yorkie (Yki), the transcriptional coactivator of the Hippo pathway, by inducing yki transcription. Moreover, both Hh signaling and Yki positively regulate the rate of FSC proliferation, both are essential for FSC maintenance, and both promote increased FSC longevity and FSC duplication when in excess. We also found that responses to activated Yki depend on Cyclin E induction while responses to excess Hh signaling depend on Yki induction, and excess Yki can compensate for defective Hh signaling. These causal connections provide the most rigorous evidence to date that a niche signal can promote stem cell maintenance principally by stimulating stem cell proliferation.
Collapse
Affiliation(s)
- Jianhua Huang
- Department of Biological Sciences, Columbia University, New York, NY 10027
| | | |
Collapse
|
46
|
Machado-Neto JA, de Melo Campos P, Olalla Saad ST, Traina F. YAP1 expression in myelodysplastic syndromes and acute leukemias. Leuk Lymphoma 2014; 55:2413-5. [PMID: 24605912 DOI: 10.3109/10428194.2014.891028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- João Agostinho Machado-Neto
- Hematology and Hemotherapy Center, University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue , Campinas, São Paulo , Brazil
| | | | | | | |
Collapse
|
47
|
Safari S, Movafagh A, Zare-Adollahi D, Ghadiani M, Riazi-Isfahani S, Safavi-Naini N, Omrani MD. MST1/2 and YAP1 gene expression in acute myeloid leukemia. Leuk Lymphoma 2014; 55:2189-91. [PMID: 24303784 DOI: 10.3109/10428194.2013.867493] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Shamsi Safari
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences , Tehran , Islamic Republic of Iran
| | | | | | | | | | | | | |
Collapse
|
48
|
Lin AYT, Pearson BJ. Planarian yorkie/YAP functions to integrate adult stem cell proliferation, organ homeostasis and maintenance of axial patterning. Development 2014; 141:1197-208. [PMID: 24523458 DOI: 10.1242/dev.101915] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
During adult homeostasis and regeneration, the freshwater planarian must accomplish a constant balance between cell proliferation and cell death, while also maintaining proper tissue and organ size and patterning. How these ordered processes are precisely modulated remains relatively unknown. Here we show that planarians use the downstream effector of the Hippo signaling cascade, yorkie (yki; YAP in vertebrates) to control a diverse set of pleiotropic processes in organ homeostasis, stem cell regulation, regeneration and axial patterning. We show that yki functions to maintain the homeostasis of the planarian excretory (protonephridial) system and to limit stem cell proliferation, but does not affect the differentiation process or cell death. Finally, we show that Yki acts synergistically with WNT/β-catenin signaling to repress head determination by limiting the expression domains of posterior WNT genes and that of the WNT-inhibitor notum. Together, our data show that yki is a key gene in planarians that integrates stem cell proliferation control, organ homeostasis, and the spatial patterning of tissues.
Collapse
Affiliation(s)
- Alexander Y T Lin
- The Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON M5G 1X8, Canada
| | | |
Collapse
|
49
|
Shin JW, Swift J, Ivanovska I, Spinler KR, Buxboim A, Discher DE. Mechanobiology of bone marrow stem cells: from myosin-II forces to compliance of matrix and nucleus in cell forms and fates. Differentiation 2013; 86:77-86. [PMID: 23790394 PMCID: PMC3964600 DOI: 10.1016/j.diff.2013.05.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 05/01/2013] [Accepted: 05/02/2013] [Indexed: 12/22/2022]
Abstract
Adult stem cells and progenitors are of great interest for their clinical application as well as their potential to reveal deep sensitivities to microenvironmental factors. The bone marrow is a niche for at least two types of stem cells, and the prototype is the hematopoietic stem cell/progenitors (HSC/Ps), which have saved many thousands of patients for several decades now. In bone marrow, HSC/Ps interact functionally with marrow stromal cells that are often referred to as mesenchymal stem cells (MSCs) or derivatives thereof. Myosin and matrix elasticity greatly affect MSC function, and these mechanobiological factors are now being explored with HSC/Ps both in vitro and in vivo. Also emerging is a role for the nucleus as a mechanically sensitive organelle that is semi-permeable to transcription factors which are modified for nuclear entry by cytoplasmic mechanobiological pathways. Since therapies envisioned with induced pluripotent stem cells and embryonic stem cells generally involve in vitro commitment to an adult stem cell or progenitor, a very deep understanding of stem cell mechanobiology is essential to progress with these multi-potent cells.
Collapse
Affiliation(s)
- Jae-Won Shin
- Molecular and Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
50
|
The transcriptional architecture of early human hematopoiesis identifies multilevel control of lymphoid commitment. Nat Immunol 2013; 14:756-63. [PMID: 23708252 PMCID: PMC4961471 DOI: 10.1038/ni.2615] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 04/16/2013] [Indexed: 12/12/2022]
Abstract
Understanding how differentiation programs originate from within the gene expression landscape of hematopoietic stem cells (HSC) is crucial to develop new clinical therapies. We mapped the transcriptional dynamics underlying the first steps of commitment by tracking transcriptome changes in human HSC and eight early progenitor populations. Transcriptional programs are extensively shared, extend across lineage-potential boundaries, and are not strictly lineage-affiliated. Elements of stem, lymphoid and myeloid programs are retained in multi-lymphoid progenitors (MLP), reflecting a hybrid transcriptional state. Based on functional single cell analysis, BCL11A, SOX4 and TEAD1 governed transcriptional networks within MLPs, leading to B cell specification. Overall, we show that integrated transcriptome approaches can identify novel regulators of multipotency and uncover additional complexity in lymphoid commitment.
Collapse
|