101
|
Hoefner C, Muhr C, Horder H, Wiesner M, Wittmann K, Lukaszyk D, Radeloff K, Winnefeld M, Becker M, Blunk T, Bauer-Kreisel P. Human Adipose-Derived Mesenchymal Stromal/Stem Cell Spheroids Possess High Adipogenic Capacity and Acquire an Adipose Tissue-like Extracellular Matrix Pattern. Tissue Eng Part A 2020; 26:915-926. [PMID: 32070231 DOI: 10.1089/ten.tea.2019.0206] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adipose-derived mesenchymal stromal/stem cells (ASCs) represent a commonly used cell source for adipose tissue engineering. In this context, ASCs have routinely been cultured in conventional 2D culture and applied as single cell suspension for seeding onto scaffold materials or direct injection. However, this approach is associated with the loss of their intrinsic 3D microenvironment and leads to impaired regenerative capacity of the cells. Thus, the application of ASCs as self-assembled 3D spheroids with cells residing in their own matrix is an attractive alternative. However, characterization of the structural features and differentiation capacity of the spheroids is necessary to effectively apply them as building blocks in adipose tissue engineering. In this study, we focus on extracellular matrix (ECM) development in ASC spheroids, as well as adipogenic differentiation in comparison to conventional 2D culture using different induction protocols. Reproducible assembly of ASCs into spheroids was achieved within 24 h using the liquid overlay technique. Undifferentiated spheroids displayed a stromal ECM pattern, with fibronectin, collagen V, and VI as the main components. In the course of adipogenesis, a dynamic shift in the ECM composition toward an adipogenic phenotype was observed, associated with enhanced expression of laminin, collagen I, IV, V, and VI, similar to native fat. Furthermore, adipogenic differentiation was enhanced in spheroids as compared with 2D cultured cells, with the spheroids needing a distinctly shorter adipogenic stimulus to sustain adipogenesis, which was demonstrated based on analysis of triglyceride content and adipogenic marker gene expression. In summary, culturing ASCs as spheroids can enhance their adipogenic capacity and generate adipose-like microtissues, which may be a promising cell delivery strategy for adipose tissue engineering approaches. Impact statement Adipose-derived mesenchymal stromal/stem cells (ASCs) as a widely used cell source for adipose tissue engineering have been shown to be limited in their regenerative capacity when applied as single cells. As an alternative approach, the delivery as spheroids, consisting of cells in a 3D context, may be favorable. However, insights into extracellular matrix (ECM) development and efficient adipogenic differentiation are required for their effective application. In this study, we show that differentiated ASC spheroids develop an ECM, resembling native adipose tissue. Furthermore, the ASC spheroids exhibited a superior differentiation capacity as compared with conventional 2D culture, and required only a short adipogenic induction stimulus. Our results identify ASC-derived spheroids as an attractive cell delivery method for adipose tissue engineering approaches.
Collapse
Affiliation(s)
- Christiane Hoefner
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| | - Christian Muhr
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| | - Hannes Horder
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| | - Miriam Wiesner
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| | - Katharina Wittmann
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| | - Daniel Lukaszyk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| | - Katrin Radeloff
- Department of Otorhinolaryngology, University of Würzburg, Würzburg, Germany
| | | | - Matthias Becker
- Institute for Medical Radiation and Cell Research, University of Würzburg, Würzburg, Germany
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| | - Petra Bauer-Kreisel
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| |
Collapse
|
102
|
McBride WH, Schaue D. Radiation-induced tissue damage and response. J Pathol 2020; 250:647-655. [PMID: 31990369 PMCID: PMC7216989 DOI: 10.1002/path.5389] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/23/2019] [Accepted: 01/20/2020] [Indexed: 12/28/2022]
Abstract
Normal tissue responses to ionizing radiation have been a major subject for study since the discovery of X-rays at the end of the 19th century. Shortly thereafter, time-dose relationships were established for some normal tissue endpoints that led to investigations into how the size of dose per fraction and the quality of radiation affected outcome. The assessment of the radiosensitivity of bone marrow stem cells using colony-forming assays by Till and McCulloch prompted the establishment of in situ clonogenic assays for other tissues that added to the radiobiology toolbox. These clonogenic and functional endpoints enabled mathematical modeling to be performed that elucidated how tissue structure, and in particular turnover time, impacted clinically relevant fractionated radiation schedules. More recently, lineage tracing technology, advanced imaging and single cell sequencing have shed further light on the behavior of cells within stem, and other, cellular compartments, both in homeostasis and after radiation damage. The discovery of heterogeneity within the stem cell compartment and plasticity in response to injury have added new dimensions to the consideration of radiation-induced tissue damage. Clinically, radiobiology of the 20th century garnered wisdom relevant to photon treatments delivered to a fairly wide field at around 2 Gy per fraction, 5 days per week, for 5-7 weeks. Recently, the scope of radiobiology has been extended by advances in technology, imaging and computing, as well as by the use of charged particles. These allow radiation to be delivered more precisely to tumors while minimizing the amount of normal tissue receiving high doses. One result has been an increase in the use of schedules with higher doses per fraction given in a shorter time frame (hypofractionation). We are unable to cover these new technologies in detail in this review, just as we must omit low-dose stochastic effects, and many aspects of dose, dose rate and radiation quality. We argue that structural diversity and plasticity within tissue compartments provides a general context for discussion of most radiation responses, while acknowledging many omissions. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- William H McBride
- Departent of Radiation OncologyUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
| | - Dörthe Schaue
- Departent of Radiation OncologyUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
| |
Collapse
|
103
|
Wang Y, Lo WC, Chou CS. Modelling stem cell ageing: a multi-compartment continuum approach. ROYAL SOCIETY OPEN SCIENCE 2020; 7:191848. [PMID: 32269805 PMCID: PMC7137970 DOI: 10.1098/rsos.191848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 02/10/2020] [Indexed: 06/11/2023]
Abstract
Stem cells are important to generate all specialized tissues at an early life stage, and in some systems, they also have repair functions to replenish the adult tissues. Repeated cell divisions lead to the accumulation of molecular damage in stem cells, which are commonly recognized as drivers of ageing. In this paper, a novel model is proposed to integrate stem cell proliferation and differentiation with damage accumulation in the stem cell ageing process. A system of two structured PDEs is used to model the population densities of stem cells (including all multiple progenitors) and terminally differentiated (TD) cells. In this system, cell cycle progression and damage accumulation are modelled by continuous dynamics, and damage segregation between daughter cells is considered at each division. Analysis and numerical simulations are conducted to study the steady-state populations and stem cell damage distributions under different damage segregation strategies. Our simulations suggest that equal distribution of the damaging substance between stem cells in a symmetric renewal and less damage retention in stem cells in the asymmetric division are favourable strategies, which reduce the death rate of the stem cells and increase the TD cell populations. Moreover, asymmetric damage segregation in stem cells leads to less concentrated damage distribution in the stem cell population, which may be more robust to the stochastic changes in the damage. The feedback regulation from stem cells can reduce oscillations and population overshoot in the process, and improve the fitness of stem cells by increasing the percentage of cells with less damage in the stem cell population.
Collapse
Affiliation(s)
- Yanli Wang
- Department of Mathematics, The Ohio State University, Columbus, OH, USA
| | - Wing-Cheong Lo
- Department of Mathematics, City University of Hong Kong, Hong Kong, People’s Republic of China
| | - Ching-Shin Chou
- Department of Mathematics, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
104
|
Clémot M, Sênos Demarco R, Jones DL. Lipid Mediated Regulation of Adult Stem Cell Behavior. Front Cell Dev Biol 2020; 8:115. [PMID: 32185173 PMCID: PMC7058546 DOI: 10.3389/fcell.2020.00115] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/11/2020] [Indexed: 12/18/2022] Open
Abstract
Adult stem cells constitute an important reservoir of self-renewing progenitor cells and are crucial for maintaining tissue and organ homeostasis. The capacity of stem cells to self-renew or differentiate can be attributed to distinct metabolic states, and it is now becoming apparent that metabolism plays instructive roles in stem cell fate decisions. Lipids are an extremely vast class of biomolecules, with essential roles in energy homeostasis, membrane structure and signaling. Imbalances in lipid homeostasis can result in lipotoxicity, cell death and diseases, such as cardiovascular disease, insulin resistance and diabetes, autoimmune disorders and cancer. Therefore, understanding how lipid metabolism affects stem cell behavior offers promising perspectives for the development of novel approaches to control stem cell behavior either in vitro or in patients, by modulating lipid metabolic pathways pharmacologically or through diet. In this review, we will first address how recent progress in lipidomics has created new opportunities to uncover stem-cell specific lipidomes. In addition, genetic and/or pharmacological modulation of lipid metabolism have shown the involvement of specific pathways, such as fatty acid oxidation (FAO), in regulating adult stem cell behavior. We will describe and compare findings obtained in multiple stem cell models in order to provide an assessment on whether unique lipid metabolic pathways may commonly regulate stem cell behavior. We will then review characterized and potential molecular mechanisms through which lipids can affect stem cell-specific properties, including self-renewal, differentiation potential or interaction with the niche. Finally, we aim to summarize the current knowledge of how alterations in lipid homeostasis that occur as a consequence of changes in diet, aging or disease can impact stem cells and, consequently, tissue homeostasis and repair.
Collapse
Affiliation(s)
- Marie Clémot
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rafael Sênos Demarco
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - D. Leanne Jones
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
105
|
Kronemberger GS, Matsui RAM, Miranda GDASDCE, Granjeiro JM, Baptista LS. Cartilage and bone tissue engineering using adipose stromal/stem cells spheroids as building blocks. World J Stem Cells 2020; 12:110-122. [PMID: 32184936 PMCID: PMC7062040 DOI: 10.4252/wjsc.v12.i2.110] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/19/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023] Open
Abstract
Scaffold-free techniques in the developmental tissue engineering area are designed to mimic in vivo embryonic processes with the aim of biofabricating, in vitro, tissues with more authentic properties. Cell clusters called spheroids are the basis for scaffold-free tissue engineering. In this review, we explore the use of spheroids from adult mesenchymal stem/stromal cells as a model in the developmental engineering area in order to mimic the developmental stages of cartilage and bone tissues. Spheroids from adult mesenchymal stromal/stem cells lineages recapitulate crucial events in bone and cartilage formation during embryogenesis, and are capable of spontaneously fusing to other spheroids, making them ideal building blocks for bone and cartilage tissue engineering. Here, we discuss data from ours and other labs on the use of adipose stromal/stem cell spheroids in chondrogenesis and osteogenesis in vitro. Overall, recent studies support the notion that spheroids are ideal "building blocks" for tissue engineering by “bottom-up” approaches, which are based on tissue assembly by advanced techniques such as three-dimensional bioprinting. Further studies on the cellular and molecular mechanisms that orchestrate spheroid fusion are now crucial to support continued development of bottom-up tissue engineering approaches such as three-dimensional bioprinting.
Collapse
Affiliation(s)
- Gabriela S Kronemberger
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, RJ 25250-020, Brazil
| | - Renata Akemi Morais Matsui
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
| | - Guilherme de Almeida Santos de Castro e Miranda
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Federal University of Rio de Janeiro (UFRJ), Campus Duque de Caxias, Duque de Caxias, RJ 25250-020, Brazil
| | - José Mauro Granjeiro
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Laboratory of Clinical Research in Odontology, Fluminense Federal University (UFF), Niterói 25255-030 Brazil
| | - Leandra Santos Baptista
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Multidisciplinary Center for Biological Research (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ) Campus Duque de Caxias, Duque de Caxias, RJ 25245-390, Brazil
| |
Collapse
|
106
|
Andrejew R, Glaser T, Oliveira-Giacomelli Á, Ribeiro D, Godoy M, Granato A, Ulrich H. Targeting Purinergic Signaling and Cell Therapy in Cardiovascular and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:275-353. [PMID: 31898792 DOI: 10.1007/978-3-030-31206-0_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular purines exert several functions in physiological and pathophysiological mechanisms. ATP acts through P2 receptors as a neurotransmitter and neuromodulator and modulates heart contractility, while adenosine participates in neurotransmission, blood pressure, and many other mechanisms. Because of their capability to differentiate into mature cell types, they provide a unique therapeutic strategy for regenerating damaged tissue, such as in cardiovascular and neurodegenerative diseases. Purinergic signaling is pivotal for controlling stem cell differentiation and phenotype determination. Proliferation, differentiation, and apoptosis of stem cells of various origins are regulated by purinergic receptors. In this chapter, we selected neurodegenerative and cardiovascular diseases with clinical trials using cell therapy and purinergic receptor targeting. We discuss these approaches as therapeutic alternatives to neurodegenerative and cardiovascular diseases. For instance, promising results were demonstrated in the utilization of mesenchymal stem cells and bone marrow mononuclear cells in vascular regeneration. Regarding neurodegenerative diseases, in general, P2X7 and A2A receptors mostly worsen the degenerative state. Stem cell-based therapy, mainly through mesenchymal and hematopoietic stem cells, showed promising results in improving symptoms caused by neurodegeneration. We propose that purinergic receptor activity regulation combined with stem cells could enhance proliferative and differentiation rates as well as cell engraftment.
Collapse
Affiliation(s)
- Roberta Andrejew
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Talita Glaser
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Ágatha Oliveira-Giacomelli
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Deidiane Ribeiro
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Mariana Godoy
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.,Laboratory of Neurodegenerative Diseases, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro Granato
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
107
|
Farzaneh M, Derakhshan Z, Hallajzadeh J, Sarani NH, Nejabatdoust A, Khoshnam SE. Suppression of TGF-β and ERK Signaling Pathways as a New Strategy to Provide Rodent and Non-Rodent Pluripotent Stem Cells. Curr Stem Cell Res Ther 2020; 14:466-473. [PMID: 30868962 DOI: 10.2174/1871527318666190314110529] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 02/02/2019] [Accepted: 02/21/2019] [Indexed: 01/07/2023]
Abstract
Stem cells are unspecialized cells and excellent model in developmental biology and a promising approach to the treatment of disease and injury. In the last 30 years, pluripotent embryonic stem (ES) cells were established from murine and primate sources, and display indefinite replicative potential and the ability to differentiate to all three embryonic germ layers. Despite large efforts in many aspects of rodent and non-rodent pluripotent stem cell culture, a number of diverse challenges remain. Natural and synthetic small molecules (SMs) strategy has the potential to overcome these hurdles. Small molecules are typically fast and reversible that target specific signaling pathways, epigenetic processes and other cellular processes. Inhibition of the transforming growth factor-β (TGF-β/Smad) and fibroblast growth factor 4 (FGF4)/ERK signaling pathways by SB431542 and PD0325901 small molecules, respectively, known as R2i, enhances the efficiency of mouse, rat, and chicken pluripotent stem cells passaging from different genetic backgrounds. Therefore, the application of SM inhibitors of TGF-β and ERK1/2 with leukemia inhibitory factor (LIF) allows the cultivation of pluripotent stem cells in a chemically defined condition. In this review, we discuss recently emerging evidence that dual inhibition of TGF-β and FGF signaling pathways plays an important role in regulating pluripotency in both rodent and non-rodent pluripotent stem cells.
Collapse
Affiliation(s)
- Maryam Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Derakhshan
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jamal Hallajzadeh
- Department of Biochemistry and Toxicology, Maraghe University of Medical Science, Maraghe, Iran
| | | | - Armin Nejabatdoust
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran
| | - Seyed Esmaeil Khoshnam
- Physiology Research Center, Department of Physiology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
108
|
Rust K, Nystul T. Signal transduction in the early Drosophila follicle stem cell lineage. CURRENT OPINION IN INSECT SCIENCE 2020; 37:39-48. [PMID: 32087562 PMCID: PMC7155752 DOI: 10.1016/j.cois.2019.11.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/13/2019] [Accepted: 11/16/2019] [Indexed: 05/08/2023]
Abstract
The follicle stem cell (FSC) lineage in the Drosophila ovary is a highly informative model of in vivo epithelial stem cell biology. Studies over the past 30 years have identified roles for every major signaling pathway in the early FSC lineage. These pathways regulate a wide variety of cell behaviors, including self-renewal, proliferation, survival and differentiation. Studies of cell signaling in the follicle epithelium have provided new insights into how these cell behaviors are coordinated within an epithelial stem cell lineage and how signaling pathways interact with each other in the native, in vivo context of a living tissue. Here, we review these studies, with a particular focus on how these pathways specify differences between the FSCs and their daughter cells. We also describe common themes that have emerged from these studies, and highlight new research directions that have been made possible by the detailed understanding of the follicle epithelium.
Collapse
|
109
|
Manini P, Lucci V, Lino V, Sartini S, Rossella F, Falco G, Chiappe C, d’Ischia M. Synthetic mycomelanin thin films as emergent bio-inspired interfaces controlling the fate of embryonic stem cells. J Mater Chem B 2020; 8:4412-4418. [DOI: 10.1039/d0tb00623h] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mycomelanin thin films from 1,8-dihydroxynaphthalene can serve as a biointerface inducing adhesion and proliferation of ESCs and promoting their differentiation towards endodermal lineages.
Collapse
Affiliation(s)
- Paola Manini
- Department of Chemical Sciences
- University of Napoli Federico II
- Complesso Universitario Monte S. Angelo
- I-80126 Napoli
- Italy
| | - Valeria Lucci
- Department of Biology
- University of Napoli Federico II
- Complesso Universitario Monte S. Angelo
- I-80126 Napoli
- Italy
| | - Valeria Lino
- Department of Chemical Sciences
- University of Napoli Federico II
- Complesso Universitario Monte S. Angelo
- I-80126 Napoli
- Italy
| | | | - Francesco Rossella
- NEST
- Scuola Normale Superiore and Istituto Nanoscienze – CNR
- I-56127 Pisa
- Italy
| | - Geppino Falco
- Department of Biology
- University of Napoli Federico II
- Complesso Universitario Monte S. Angelo
- I-80126 Napoli
- Italy
| | - Cinzia Chiappe
- Department of Pharmacy
- University of Pisa
- I-56126 Pisa
- Italy
| | - Marco d’Ischia
- Department of Chemical Sciences
- University of Napoli Federico II
- Complesso Universitario Monte S. Angelo
- I-80126 Napoli
- Italy
| |
Collapse
|
110
|
An S, Han SY, Cho SW. Hydrogel-integrated Microfluidic Systems for Advanced Stem Cell Engineering. BIOCHIP JOURNAL 2019. [DOI: 10.1007/s13206-019-3402-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
111
|
Guan S, Zhang K, Li J. Recent Advances in Extracellular Matrix for Engineering Stem Cell Responses. Curr Med Chem 2019; 26:6321-6338. [DOI: 10.2174/0929867326666190704121309] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/02/2018] [Accepted: 01/25/2019] [Indexed: 02/06/2023]
Abstract
Stem cell transplantation is an advanced medical technology, which brings hope for the
treatment of some difficult diseases in the clinic. Attributed to its self-renewal and differential
ability, stem cell research has been pushed to the forefront of regenerative medicine and has become
a hot topic in tissue engineering. The surrounding extracellular matrix has physical functions
and important biological significance in regulating the life activities of cells, which may play crucial
roles for in situ inducing specific differentiation of stem cells. In this review, we discuss the
stem cells and their engineering application, and highlight the control of the fate of stem cells, we
offer our perspectives on the various challenges and opportunities facing the use of the components
of extracellular matrix for stem cell attachment, growth, proliferation, migration and differentiation.
Collapse
Affiliation(s)
- Shuaimeng Guan
- School of Life Science, Zhengzhou University, Zhengzhou 450000, China
| | - Kun Zhang
- School of Life Science, Zhengzhou University, Zhengzhou 450000, China
| | - Jingan Li
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450000, China
| |
Collapse
|
112
|
La HM, Hobbs RM. Mechanisms regulating mammalian spermatogenesis and fertility recovery following germ cell depletion. Cell Mol Life Sci 2019; 76:4071-4102. [PMID: 31254043 PMCID: PMC11105665 DOI: 10.1007/s00018-019-03201-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 06/07/2019] [Accepted: 06/19/2019] [Indexed: 12/19/2022]
Abstract
Mammalian spermatogenesis is a highly complex multi-step process sustained by a population of mitotic germ cells with self-renewal potential known as spermatogonial stem cells (SSCs). The maintenance and regulation of SSC function are strictly dependent on a supportive niche that is composed of multiple cell types. A detailed appreciation of the molecular mechanisms underpinning SSC activity and fate is of fundamental importance for spermatogenesis and male fertility. However, different models of SSC identity and spermatogonial hierarchy have been proposed and recent studies indicate that cell populations supporting steady-state germline maintenance and regeneration following damage are distinct. Importantly, dynamic changes in niche properties may underlie the fate plasticity of spermatogonia evident during testis regeneration. While formation of spermatogenic colonies in germ-cell-depleted testis upon transplantation is a standard assay for SSCs, differentiation-primed spermatogonial fractions have transplantation potential and this assay provides readout of regenerative rather than steady-state stem cell capacity. The characterisation of spermatogonial populations with regenerative capacity is essential for the development of clinical applications aimed at restoring fertility in individuals following germline depletion by genotoxic treatments. This review will discuss regulatory mechanisms of SSCs in homeostatic and regenerative testis and the conservation of these mechanisms between rodent models and man.
Collapse
Affiliation(s)
- Hue M La
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, 3800, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Robin M Hobbs
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, 3800, Australia.
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia.
| |
Collapse
|
113
|
Kitade M, Kaji K, Nishimura N, Seki K, Nakanishi K, Tsuji Y, Sato S, Saikawa S, Takaya H, Kawaratani H, Namisaki T, Moriya K, Mitoro A, Yoshiji H. Blocking development of liver fibrosis augments hepatic progenitor cell-derived liver regeneration in a mouse chronic liver injury model. Hepatol Res 2019; 49:1034-1045. [PMID: 30989766 DOI: 10.1111/hepr.13351] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 02/08/2023]
Abstract
AIM The roles of hepatic progenitor cells (HPCs) in regeneration of a diseased liver are unclear. Hepatic stellate cells (HSCs) contribute to liver fibrosis but are also a component of the HPC niche. Hepatic progenitor cells expand along with HSC activation and liver fibrosis. However, little is known about the interplay of liver fibrosis and HPC-mediated liver regeneration. This study aimed to investigate HSCs and HPCs in liver regeneration. METHODS Liver injury in mice was induced with 3,5-diethoxycarbonyl-1,4-dihydrocollidine, and HPC expansion and fibrosis were assessed. An angiotensin II type 1 receptor blocker (ARB) was administered to assess its effect on fibrosis and regeneration. RESULTS Treatment with ARB attenuated fibrosis and expansion of α-smooth muscle actin-positive activated HSCs as indicated by increased liver weight and Ki-67-positive hepatocytes. Immunohistochemical staining suggested that HPC differentiation was shifted toward hepatocytes (HCs) when ARB treatment decreased HPC encapsulation by HSCs and extracellular matrix. Conditioned medium produced by culturing the human HSC LX-2 line strongly augmented differentiation to biliary epithelial cells (BECs) but inhibited that to HCs. Activated HSCs expressed Jagged1, a NOTCH ligand, which plays a central role in differentiation of HPCs toward BECs. CONCLUSIONS Hepatic stellate cells, the HPC niche cells, control differentiation of HPCs, directing them toward BECs rather than HCs in a diseased liver model. Antifibrosis treatment with an ARB preferentially redirects HPC differentiation toward HCs by blocking the NOTCH pathway in the HPC niche, resulting in more efficient HPC-mediated liver regeneration.
Collapse
Affiliation(s)
- Mitsuteru Kitade
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| | - Kosuke Kaji
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| | - Norihisa Nishimura
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| | - Kenichiro Seki
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| | - Keisuke Nakanishi
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| | - Yuki Tsuji
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| | - Shinya Sato
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| | - Soichiro Saikawa
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| | - Hiroaki Takaya
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| | - Hideto Kawaratani
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| | - Tadashi Namisaki
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| | - Kei Moriya
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| | - Akira Mitoro
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| | - Hitoshi Yoshiji
- Third Department of Internal Medicine, Nara Medical University, Nara, Japan
| |
Collapse
|
114
|
Das D, Fletcher RB, Ngai J. Cellular mechanisms of epithelial stem cell self-renewal and differentiation during homeostasis and repair. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e361. [PMID: 31468728 DOI: 10.1002/wdev.361] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 12/14/2022]
Abstract
Epithelia in adult mammals exhibit remarkable regenerative capacities owing to the presence of adult stem cells, which self-renew and differentiate to replace cells lost to normal turnover or injury. The mechanisms supporting tissue homeostasis and injury-induced repair often differ from each other as well as from those used in embryonic development. Recent studies have also highlighted the phenomenon of cellular plasticity in adult tissues, in which differentiated cells can change fate and even give rise to new stem cell populations to complement the canonical stem cells in promoting repair following injury. Signaling pathways such as WNT, bone morphogenetic protein, and Sonic Hedgehog play critical roles in stem cell maintenance and cell fate decisions across diverse epithelia and conditions, suggesting that conserved mechanisms underlie the regenerative capacity of adult epithelial structures. This article is categorized under: Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Adult Stem Cells, Tissue Renewal, and Regeneration > Tissue Stem Cells and Niches Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cell Differentiation and Reversion Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration.
Collapse
Affiliation(s)
- Diya Das
- Department of Molecular and Cell Biology, University of California, Berkeley, California.,Berkeley Institute for Data Science, University of California, Berkeley, California
| | - Russell B Fletcher
- Department of Molecular and Cell Biology, University of California, Berkeley, California
| | - John Ngai
- Department of Molecular and Cell Biology, University of California, Berkeley, California.,Helen Wills Neuroscience Institute, University of California, Berkeley, California.,QB3 Functional Genomics Laboratory, University of California, Berkeley, California
| |
Collapse
|
115
|
A stochastic model of adult neurogenesis coupling cell cycle progression and differentiation. J Theor Biol 2019; 475:60-72. [PMID: 31128140 DOI: 10.1016/j.jtbi.2019.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 05/16/2019] [Accepted: 05/22/2019] [Indexed: 11/22/2022]
Abstract
Long-term tissue homeostasis requires a precise balance between stem cell self-renewal and the generation of differentiated progeny. Recently, it has been shown that in the adult murine brain, neural stem cells (NSCs) divide mostly symmetrically. This finding suggests that the required balance for tissue homeostasis is accomplished at the population level. However, it remains unclear how this balance is enabled. Furthermore, there is experimental evidence that proneural differentiation factors not only promote differentiation, but also cell cycle progression, suggesting a link between the two processes in NSCs. To study the effect of such a link on NSC dynamics, we developed a stochastic model in which stem cells have an intrinsic probability to progress through cell cycle and to differentiate. Our results show that increasing heterogeneity in differentiation probabilities leads to a decreased probability of long-term tissue homeostasis, and that this effect can be compensated when cell cycle progression and differentiation are positively coupled. Using single-cell RNA-Seq profiling of adult NSCs, we found a positive correlation in the expression levels of cell cycle and differentiation markers. Our findings suggest that a coupling between cell cycle progression and differentiation on the cellular level is part of the process that maintains tissue homeostasis in the adult brain.
Collapse
|
116
|
Stem cell-based bone and dental regeneration: a view of microenvironmental modulation. Int J Oral Sci 2019; 11:23. [PMID: 31423011 PMCID: PMC6802669 DOI: 10.1038/s41368-019-0060-3] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/28/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023] Open
Abstract
In modern medicine, bone and dental loss and defects are common and widespread morbidities, for which regenerative therapy has shown great promise. Mesenchymal stem cells, obtained from various sources and playing an essential role in organ development and postnatal repair, have exhibited enormous potential for regenerating bone and dental tissue. Currently, mesenchymal stem cells (MSCs)-based bone and dental regeneration mainly includes two strategies: the rescue or mobilization of endogenous MSCs and the application of exogenous MSCs in cytotherapy or tissue engineering. Nevertheless, the efficacy of MSC-based regeneration is not always fulfilled, especially in diseased microenvironments. Specifically, the diseased microenvironment not only impairs the regenerative potential of resident MSCs but also controls the therapeutic efficacy of exogenous MSCs, both as donors and recipients. Accordingly, approaches targeting a diseased microenvironment have been established, including improving the diseased niche to restore endogenous MSCs, enhancing MSC resistance to a diseased microenvironment and renormalizing the microenvironment to guarantee MSC-mediated therapies. Moreover, the application of extracellular vesicles (EVs) as cell-free therapy has emerged as a promising therapeutic strategy. In this review, we summarize current knowledge regarding the tactics of MSC-based bone and dental regeneration and the decisive role of the microenvironment, emphasizing the therapeutic potential of microenvironment-targeting strategies in bone and dental regenerative medicine.
Collapse
|
117
|
Ma L, Hu J, Cao Y, Xie Y, Wang H, Fan Z, Zhang C, Wang J, Wu CT, Wang S. Maintained Properties of Aged Dental Pulp Stem Cells for Superior Periodontal Tissue Regeneration. Aging Dis 2019; 10:793-806. [PMID: 31440385 PMCID: PMC6675537 DOI: 10.14336/ad.2018.0729] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/29/2018] [Indexed: 12/23/2022] Open
Abstract
Owing to excellent therapeutic potential, mesenchymal stem cells (MSCs) are gaining increasing popularity with researchers worldwide for applications in tissue engineering, and in treatment of inflammation-related and age-related disorders. However, the senescence of MSCs over passaging has limited their clinical application owing to adverse effect on physiological function maintenance of tissues as well as disease treatment. An inflammatory microenvironment is one of the key contributors to MSC senescence, resulting in low regeneration efficiency. Therefore, MSCs with high resistance to cellular senescence would be a benefit for tissue regeneration. Toward this end, we analyzed the senescence properties of different types of stem cells during culture and under inflammation, including dental pulp stem cells (DPSCs), periodontal ligament stem cells (PDLSCs), bone marrow mesenchymal stem cells (BMMSCs), and adipose-derived stem cells (ADSCs). Overall, the DPSCs had higher proliferation rates, lower cellular senescence, and enhanced osteogenesis maintenance compared to those of non-dental MSCs cultured from passage three to six. The expression profiles of genes related to apoptosis, cell cycle, and cellular protein metabolic process (contributing to the cell self-renewal ability and metabolic processes) significantly differed between DPSCs and BMMSCs at passage three. Moreover, DPSCs were superior to BMMSCs with regards to resistance to lipopolysaccharide-induced apoptosis and senescence, with enhanced osteogenesis in vitro, and showed improved periodontal regeneration after injection in a miniature pig periodontitis model in vivo. Overall, the present study indicates that DPSCs show superior resistance to subculture and inflammation-induced senescence and would be suitable stem cells for tissue engineering with inflammation.
Collapse
Affiliation(s)
- Linsha Ma
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China
| | - Jingchao Hu
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China
| | - Yu Cao
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China
| | - Yilin Xie
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China
| | - Hua Wang
- 2Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhipeng Fan
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China
| | - Chunmei Zhang
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China
| | - Jinsong Wang
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China.,3Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Chu-Tse Wu
- 2Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Songlin Wang
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China.,3Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| |
Collapse
|
118
|
Sasikumar S, Chameettachal S, Cromer B, Pati F, Kingshott P. Decellularized extracellular matrix hydrogels—cell behavior as a function of matrix stiffness. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019. [DOI: 10.1016/j.cobme.2019.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
119
|
Al-Hazmi N, Alhazzazi T, Williams G, Stoeber K, Al-Dabbagh R. DNA replication licensing factor MCM2, geminin, and Ki67 define proliferative state and are linked with survival in oral squamous cell carcinoma. Eur J Oral Sci 2019; 126:186-196. [PMID: 29745471 DOI: 10.1111/eos.12420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Oral squamous cell carcinoma (OSCC) is still an unabated global killer with little advancement in its survival rate. DNA replication licensing proteins and Aurora kinase A are biomarkers that play important roles in genomic stability. The expression profile of minichromosomal maintenance protein 2 (MCM2), Ki67, geminin, and Aurora-A were linked to clinicopathological and outcome parameters, survival, and DNA content in 125 cases of OSCC. Oral fibroepithelial polyps (OFEP) were controls. The OSCC tumour cells were in a rapidly proliferating state, as assessed by the increased expression profile of MCM2, Ki67, geminin, and Aurora-A and of the geminin/Ki67 ratio, and the decrease of the MCM2/Ki67 ratio, in OSCC compared with OFEP (P < 0.000). There was an association between expression of MCM2, Ki67, and geminin and tumour histologic and invasive front grade (P < 0.05). A total of 82% of the OSCC assessed had aneuploid DNA content, which was associated with increased expression intensity of Aurora-A (P = 0.01). Geminin and the geminin/Ki67 ratio were associated with TNM staging (P < 0.05), and weak expression of MCM2, Ki67, geminin, and Aurora-A were predictive of OSCC survival (P < 0.05). Dysregulation of the origin licensing pathway and the mitotic pathway are important events in OSCC, and the combined analysis of these proteins may contribute to improved treatment decisions.
Collapse
Affiliation(s)
- Nadia Al-Hazmi
- Department of Oral Biology, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Turki Alhazzazi
- Department of Oral Biology, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Gareth Williams
- Oncologica, London, UK.,Department of Pathology, UCL Cancer Institute, University College London, London, UK
| | - Kai Stoeber
- Department of Pathology, UCL Cancer Institute, University College London, London, UK
| | - Raghad Al-Dabbagh
- Department of Oral and Maxillofacial Prosthodontics, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
120
|
Mitchell R, Mellows B, Sheard J, Antonioli M, Kretz O, Chambers D, Zeuner MT, Tomkins JE, Denecke B, Musante L, Joch B, Debacq-Chainiaux F, Holthofer H, Ray S, Huber TB, Dengjel J, De Coppi P, Widera D, Patel K. Secretome of adipose-derived mesenchymal stem cells promotes skeletal muscle regeneration through synergistic action of extracellular vesicle cargo and soluble proteins. Stem Cell Res Ther 2019; 10:116. [PMID: 30953537 PMCID: PMC6451311 DOI: 10.1186/s13287-019-1213-1] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 02/12/2019] [Accepted: 03/13/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The mechanisms underpinning the regenerative capabilities of mesenchymal stem cells (MSC) were originally thought to reside in their ability to recognise damaged tissue and to differentiate into specific cell types that would replace defective cells. However, recent work has shown that molecules produced by MSCs (secretome), particularly those packaged in extracellular vesicles (EVs), rather than the cells themselves are responsible for tissue repair. METHODS Here we have produced a secretome from adipose-derived mesenchymal stem cells (ADSC) that is free of exogenous molecules by incubation within a saline solution. Various in vitro models were used to evaluate the effects of the secretome on cellular processes that promote tissue regeneration. A cardiotoxin-induced skeletal muscle injury model was used to test the regenerative effects of the whole secretome or isolated extracellular vesicle fraction in vivo. This was followed by bioinformatic analysis of the components of the protein and miRNA content of the secretome and finally compared to a secretome generated from a secondary stem cell source. RESULTS Here we have demonstrated that the secretome from adipose-derived mesenchymal stem cells shows robust effects on cellular processes that promote tissue regeneration. Furthermore, we show that the whole ADSC secretome is capable of enhancing the rate of skeletal muscle regeneration following acute damage. We assessed the efficacy of the total secretome compared with the extracellular vesicle fraction on a number of assays that inform on tissue regeneration and demonstrate that both fractions affect different aspects of the process in vitro and in vivo. Our in vitro, in vivo, and bioinformatic results show that factors that promote regeneration are distributed both within extracellular vesicles and the soluble fraction of the secretome. CONCLUSIONS Taken together, our study implies that extracellular vesicles and soluble molecules within ADSC secretome act in a synergistic manner to promote muscle generation.
Collapse
Affiliation(s)
- Robert Mitchell
- School of Biological Sciences, University of Reading, Reading, UK
| | - Ben Mellows
- School of Biological Sciences, University of Reading, Reading, UK
| | - Jonathan Sheard
- Stem Cell Biology and Regenerative Biology Group, School of Pharmacy, University of Reading, Reading, UK
- Sheard BioTech Ltd, 20-22 Wenlock Road, London, N1 7GU UK
| | | | - Oliver Kretz
- Department of Medicine III, Faculty of Medicine University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Renal Division, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - David Chambers
- Wolfson Centre for Age-Related Diseases, King’s College, London, UK
| | - Marie-Theres Zeuner
- Stem Cell Biology and Regenerative Biology Group, School of Pharmacy, University of Reading, Reading, UK
| | - James E. Tomkins
- Stem Cell Biology and Regenerative Biology Group, School of Pharmacy, University of Reading, Reading, UK
| | - Bernd Denecke
- Interdisciplinary Centre for Clinical Research Aachen, RWTH Aachen University, Aachen, Germany
| | - Luca Musante
- Centre for Bioanalytical Sciences (CBAS), Dublin City University, Dublin, Ireland
| | - Barbara Joch
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Harry Holthofer
- Centre for Bioanalytical Sciences (CBAS), Dublin City University, Dublin, Ireland
- FRIAS Freiburg Institute for Advanced Studies, University of Freiburg, Freiburg, Germany
| | - Steve Ray
- Micregen, Alderley Edge, Manchester, UK
| | - Tobias B. Huber
- Department of Medicine III, Faculty of Medicine University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Renal Division, Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- FRIAS Freiburg Institute for Advanced Studies, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies and Centre for Systems Biology (ZBSA), Albert-Ludwigs-University, Freiburg, Germany
| | - Joern Dengjel
- FRIAS Freiburg Institute for Advanced Studies, University of Freiburg, Freiburg, Germany
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Paolo De Coppi
- Stem Cells & Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Darius Widera
- Stem Cell Biology and Regenerative Biology Group, School of Pharmacy, University of Reading, Reading, UK
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK
- FRIAS Freiburg Institute for Advanced Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
121
|
Brown Y, Hua S, Tanwar PS. Extracellular matrix-mediated regulation of cancer stem cells and chemoresistance. Int J Biochem Cell Biol 2019; 109:90-104. [DOI: 10.1016/j.biocel.2019.02.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 02/03/2019] [Accepted: 02/05/2019] [Indexed: 12/12/2022]
|
122
|
Hellmund KS, Koksch B. Self-Assembling Peptides as Extracellular Matrix Mimics to Influence Stem Cell's Fate. Front Chem 2019; 7:172. [PMID: 31001512 PMCID: PMC6455064 DOI: 10.3389/fchem.2019.00172] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/05/2019] [Indexed: 12/13/2022] Open
Abstract
Interest in biologically active materials that can be used as cell culture substrates for medicinal applications has increased dramatically over the last decade. The design and development of biomaterials mimicking the natural environment of different cell types, the so-called extracellular matrix (ECM), is the focus of research in this field. The ECM exists as an ensemble of several adhesion proteins with different functionalities that can be presented to the embedded cells. These functionalities regulate numerous cellular processes. Therefore, different approaches and strategies using peptide- and protein-based biopolymers have been investigated to support the proliferation, differentiation, and self-renewal of stem cells, in the context of regenerative medicine. This minireview summarizes recent developments in this area, with a focus on peptide-based biomaterials used as stem cell culture substrates.
Collapse
Affiliation(s)
| | - Beate Koksch
- Department of Biology, Chemistry, and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
123
|
Tsingos E, Höckendorf B, Sütterlin T, Kirchmaier S, Grabe N, Centanin L, Wittbrodt J. Retinal stem cells modulate proliferative parameters to coordinate post-embryonic morphogenesis in the eye of fish. eLife 2019; 8:42646. [PMID: 30910010 PMCID: PMC6486154 DOI: 10.7554/elife.42646] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 03/13/2019] [Indexed: 12/22/2022] Open
Abstract
Combining clonal analysis with a computational agent based model, we investigate how tissue-specific stem cells for neural retina (NR) and retinal pigmented epithelium (RPE) of the teleost medaka (Oryzias latipes) coordinate their growth rates. NR cell division timing is less variable, consistent with an upstream role as growth inducer. RPE cells divide with greater variability, consistent with a downstream role responding to inductive signals. Strikingly, the arrangement of the retinal ciliary marginal zone niche results in a spatially biased random lineage loss, where stem- and progenitor cell domains emerge spontaneously. Further, our data indicate that NR cells orient division axes to regulate organ shape and retinal topology. We highlight an unappreciated mechanism for growth coordination, where one tissue integrates cues to synchronize growth of nearby tissues. This strategy may enable evolution to modulate cell proliferation parameters in one tissue to adapt whole-organ morphogenesis in a complex vertebrate organ. By the time babies reach adulthood, they have grown many times larger than they were at birth. This development is driven by an increase in the number and size of cells in the body. In particular, special types of cells, called stem cells, act as a reservoir for tissues: they divide to create new cells that will mature into various specialized structures. The retina is the light-sensitive part of the eye. It consists of the neural retina, a tissue that contains light-detecting cells, which is supported by the retinal pigment epithelium or RPE. In fish, the RPE and neural retina are replenished by distinct groups of stem cells that do not mix, despite the tissues being close together. Unlike humans, fish grow throughout adulthood, and their eyes must then keep pace with the body. This means that the different tissues in the retina must somehow coordinate to expand at the same rate: otherwise, the retina would get wrinkled and not work properly. Tsingos et al. therefore wanted to determine how stem cells in the neural retina and RPE co-operated to produce the right number of new cells at the right time. First, stem cells in the eyes of newly hatched fish were labelled with a visible marker so that their divisions could be tracked over time to build cell family trees. This showed that stem cells behaved differently in the neural retina and the RPE. Computer simulations of the growing retina explained this behavior: stem cells in the neural retina were telling the RPE stem cells when it was time to divide. Combining results from the simulations with data from the experiments revealed that a stem cell decided to keep up dividing partly because of its position in the tissue, and partly because of random chance. To be healthy, the body needs to fine-tune the number of cells it produces: creating too few cells may make it difficult to heal after injury, but making too many could lead to diseases such as cancer. Understanding how tissues normally agree to grow together could therefore open new avenues of treatment for these conditions.
Collapse
Affiliation(s)
- Erika Tsingos
- Centre for Organismal Studies Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Burkhard Höckendorf
- Centre for Organismal Studies Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Thomas Sütterlin
- National Center for Tumor Diseases, Hamamatsu TIGA Center, Bioquant, Heidelberg University, Heidelberg, Germany
| | - Stephan Kirchmaier
- Centre for Organismal Studies Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Niels Grabe
- National Center for Tumor Diseases, Hamamatsu TIGA Center, Bioquant, Heidelberg University, Heidelberg, Germany
| | - Lazaro Centanin
- Centre for Organismal Studies Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Joachim Wittbrodt
- Centre for Organismal Studies Heidelberg, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
124
|
Zhou X, Ma Y, Liu F, Gu C, Wang X, Xia H, Zhou G, Huang J, Luo X, Yang J. Melanocyte Chitosan/Gelatin Composite Fabrication with Human Outer Root Sheath-Derived Cells to Produce Pigment. Sci Rep 2019; 9:5198. [PMID: 30914712 PMCID: PMC6435804 DOI: 10.1038/s41598-019-41611-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 03/11/2019] [Indexed: 12/26/2022] Open
Abstract
The hair follicle serves as a melanocyte reservoir for both hair and skin pigmentation. Melanocyte stem cells (MelSCs) and melanocyte progenitors reside in the bulge/sub-bulge region of the lower permanent portion of the hair follicle and play a vital role for repigmentation in vitiligo. It would be beneficial to isolate MelSCs in order to further study their function in pigmentary disorders; however, due to the lack of specific molecular surface markers, this has not yet been successfully accomplished in human hair follicles (HuHF). One potential method for MelSCs isolation is the “side population” technique, which is frequently used to isolate hematopoietic and tumor stem cells. In the present study, we decided to isolate HuHF MelSCs using “side population” to investigate their melanotic function. By analyzing mRNA expression of TYR, SOX10, and MITF, melanosome structure, and immunofluorescence with melanocyte-specific markers, we revealed that the SP-fraction contained MelSCs with an admixture of differentiated melanocytes. Furthermore, our in vivo studies indicated that differentiated SP-fraction cells, when fabricated into a cell-chitosan/gelatin composite, could transiently repopulate immunologically compromised mice skin to regain pigmentation. In summary, the SP technique is capable of isolating HuHF MelSCs that can potentially be used to repopulate skin for pigmentation.
Collapse
Affiliation(s)
- Xianyu Zhou
- Department of Plastic and Reconstructive Surgery, the Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Yan Ma
- Division of Plastic Surgery, Xinjiang Korla Bazhou People's Hospital, Xinjiang, People's Republic of China
| | - Fei Liu
- Department of Plastic and Reconstructive Surgery, the Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Chuan Gu
- Department of Plastic and Reconstructive Surgery, the Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Xiuxia Wang
- Department of Plastic and Reconstructive Surgery, the Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Huitang Xia
- Department of Plastic and Reconstructive Surgery, the Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, the Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Jinny Huang
- Department of Transplantation, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xusong Luo
- Department of Plastic and Reconstructive Surgery, the Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China.
| | - Jun Yang
- Department of Plastic and Reconstructive Surgery, the Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
125
|
Sánchez-Romero N, Sainz-Arnal P, Pla-Palacín I, Dachary PR, Almeida H, Pastor C, Soto DR, Rodriguez MC, Arbizu EO, Martinez LB, Serrano-Aulló T, Baptista PM. The role of extracellular matrix on liver stem cell fate: A dynamic relationship in health and disease. Differentiation 2019; 106:49-56. [PMID: 30878881 DOI: 10.1016/j.diff.2019.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 02/07/2023]
Abstract
The liver stem cell niche is a specialized and dynamic microenvironment with biomechanical and biochemical characteristics that regulate stem cell behavior. This is feasible due to the coordination of a complex network of secreted factors, small molecules, neural, blood inputs and extracellular matrix (ECM) components involved in the regulation of stem cell fate (self-renewal, survival, and differentiation into more mature phenotypes like hepatocytes and cholangiocytes). In this review, we describe and summarize all the major components that play essential roles in the liver stem cell niche, in particular, growth factor signaling and the biomechanical properties of the ECM.
Collapse
Affiliation(s)
| | - Pilar Sainz-Arnal
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain; Aragon's Health Science Research Institute (IACS), Zaragoza, Spain
| | - Iris Pla-Palacín
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
| | | | - Helen Almeida
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
| | - Cristina Pastor
- Aragon's Health Science Research Institute (IACS), Zaragoza, Spain
| | - Daniela Rubio Soto
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain; Health Research Institute of Jiménez Díaz Foundation (IIS FJD), Madrid, Spain; Biomedical and Aerospace Engineering Department, University Carlos III of Madrid, Spain
| | | | | | | | | | - Pedro M Baptista
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain; Health Research Institute of Jiménez Díaz Foundation (IIS FJD), Madrid, Spain; Biomedical and Aerospace Engineering Department, University Carlos III of Madrid, Spain.
| |
Collapse
|
126
|
Zhang J, Zhao J, Chen Y, Shi H, Huang X, Wang Y, Wang Y, Wei Y, Xue W, Han J. Effect of mGluR7 on proliferation of human embryonic neural stem cells. Medicine (Baltimore) 2019; 98:e14683. [PMID: 30817600 PMCID: PMC6831331 DOI: 10.1097/md.0000000000014683] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
This study is to investigate the effect of metabotropic glutamate receptor 7 (mGluR7) on the proliferation of human embryonic neural stem cells (NSCs) and its molecular mechanism.Human embryonic NSCs were isolated. The pCMV2-GV146-GFP-mGluR7 plasmid was transfected to over-express mGluR7 while mGluR7 siRNA was transfected to knockdown mGluR7. MTT assay was used to analyze cell proliferation. Flow cytometry was used to detect cell cycle and apoptosis. Protein and mRNA levels were analyzed by Western blot and RT-qPCR, respectively.The viability of human NSCs and the diameter of neurospheres after 24 hours, 48 hours, and 72 hours of transfection significantly increased by mGluR7 overexpression whereas significantly decreased by mGluR7 knockdown. Ki-67 expression was up-regulated by mGluR7 overexpression whereas down-regulated by mGluR7 siRNA, indicating a promotive effect of mGluR7 on NSC proliferation. After mGluR7 overexpression, G1/G0 phase cell ratio dropped significantly compared with control group, while the S phase cell ratio increased. mGluR7 silencing arrested human NSCs at G1/G0 phase. After 48 hours of transfection, there was a decrease of apoptosis by mGluR7 overexpression, while mGluR7 silencing induced apoptosis of human NSCs. Additionally, overexpression of mGluR7 up-regulated the expression of p-serine/threonine kinase (AKT), cyclin D1, and cyclin-dependent kinase 2 (CDK2). The mGluR7 knockdown had opposite effects. Similarly, mGluR7 down-regulated the expression of Caspase-3/9, while the mGluR7 knockdown promoted this.mGluR7 can promote the proliferation of human embryonic cortical NSCs in vitro. This effect may be mediated by promoting cell cycle progression, inhibiting cell apoptosis, activating the AKT signaling pathway, and inhibiting the Caspase-3/9 signaling pathway.
Collapse
|
127
|
Takahashi H, Sakata N, Yoshimatsu G, Hasegawa S, Kodama S. Regenerative and Transplantation Medicine: Cellular Therapy Using Adipose Tissue-Derived Mesenchymal Stromal Cells for Type 1 Diabetes Mellitus. J Clin Med 2019; 8:249. [PMID: 30781427 PMCID: PMC6406504 DOI: 10.3390/jcm8020249] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/09/2019] [Accepted: 02/13/2019] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is caused by the autoimmune targeting of pancreatic β-cells, and, in the advanced stage, severe hypoinsulinemia due to islet destruction. In patients with T1DM, continuous exogenous insulin therapy cannot be avoided. However, an insufficient dose of insulin easily induces extreme hyperglycemia or diabetic ketoacidosis, and intensive insulin therapy may cause hypoglycemic symptoms including hypoglycemic shock. While these insulin therapies are efficacious in most patients, some additional therapies are warranted to support the control of blood glucose levels and reduce the risk of hypoglycemia in patients who respond poorly despite receiving appropriate treatment. There has been a recent gain in the popularity of cellular therapies using mesenchymal stromal cells (MSCs) in various clinical fields, owing to their multipotentiality, capacity for self-renewal, and regenerative and immunomodulatory potential. In particular, adipose tissue-derived MSCs (ADMSCs) have become a focus in the clinical setting due to the abundance and easy isolation of these cells. In this review, we outline the possible therapeutic benefits of ADMSC for the treatment of T1DM.
Collapse
Affiliation(s)
- Hiroyuki Takahashi
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Department of Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Naoaki Sakata
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Gumpei Yoshimatsu
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Suguru Hasegawa
- Department of Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Shohta Kodama
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| |
Collapse
|
128
|
The monoclonal antibody EPR1614Y against the stem cell biomarker keratin K15 lacks specificity and reacts with other keratins. Sci Rep 2019; 9:1943. [PMID: 30760780 PMCID: PMC6374370 DOI: 10.1038/s41598-018-38163-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 12/20/2018] [Indexed: 12/17/2022] Open
Abstract
Keratin 15 (K15), a type I keratin, which pairs with K5 in epidermis, has been used extensively as a biomarker for stem cells. Two commercial antibodies, LHK15, a mouse monoclonal and EPR1614Y, a rabbit monoclonal, have been widely employed to study K15 expression. Here we report differential reactivity of these antibodies on epithelial cells and tissue sections. Although the two antibodies specifically recognised K15 on western blot, they reacted differently on skin sections and cell lines. LHK15 reacted in patches, whereas EPR1614Y reacted homogenously with the basal keratinocytes in skin sections. In cultured cells, LHK15 did not react with K15 deficient NEB-1, KEB-11, MCF-7 and SW13 cells expressing only exogenous K8 and K18 but reacted when these cells were transduced with K15. On the other hand, EPR1614Y reacted with these cells even though they were devoid of K15. Taken together these results suggest that EPR1614Y recognises a conformational epitope on keratin filaments which can be reconstituted by other keratins as well as by K15. In conclusion, this report highlights that all commercially available antibodies may not be equally specific in identifying the K15 positive stem cell.
Collapse
|
129
|
Yokoi M, Kuremoto KI, Okada S, Sasaki M, Tsuga K. Effect of attenuation of fibroblast growth factor receptor 2b signaling on odontoblast differentiation and dentin formation. In Vitro Cell Dev Biol Anim 2019; 55:211-219. [DOI: 10.1007/s11626-019-00323-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/16/2019] [Indexed: 12/14/2022]
|
130
|
Sousa‐Franco A, Rebelo K, da Rocha ST, Bernardes de Jesus B. LncRNAs regulating stemness in aging. Aging Cell 2019; 18:e12870. [PMID: 30456884 PMCID: PMC6351848 DOI: 10.1111/acel.12870] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 09/18/2018] [Accepted: 09/28/2018] [Indexed: 12/21/2022] Open
Abstract
One of the most outstanding observations from next-generation sequencing approaches was that only 1.5% of our genes code for proteins. The biggest part is transcribed but give rise to different families of RNAs without coding potential. The functional relevance of these abundant transcripts remains far from elucidated. Among them are the long non-coding RNAs (lncRNAs), a relatively large and heterogeneous group of RNAs shown to be highly tissue-specific, indicating a prominent role in processes controlling cellular identity. In particular, lncRNAs have been linked to both stemness properties and detrimental pathways regulating the aging process, being novel players in the intricate network guiding tissue homeostasis. Here, we summarize the up-to-date information on the role of lncRNAs that affect stemness and hence impact upon aging, highlighting the likelihood that lncRNAs may represent an unexploited reservoir of potential therapeutic targets for reprogramming applications and aging-related diseases.
Collapse
Affiliation(s)
- António Sousa‐Franco
- Instituto de Medicina MolecularFaculdade de Medicina da Universidade de LisboaLisboaPortugal
| | - Kenny Rebelo
- Instituto de Medicina MolecularFaculdade de Medicina da Universidade de LisboaLisboaPortugal
| | - Simão Teixeira da Rocha
- Instituto de Medicina MolecularFaculdade de Medicina da Universidade de LisboaLisboaPortugal
| | - Bruno Bernardes de Jesus
- Instituto de Medicina MolecularFaculdade de Medicina da Universidade de LisboaLisboaPortugal
- Department of Medical Sciences and Institute of Biomedicine—iBiMEDUniversity of AveiroAveiroPortugal
| |
Collapse
|
131
|
Abstract
A multilayered epithelium to fulfil its function must be replaced throughout the lifespan. This is possible due to the presence of multipotent, self-renewing epidermal stem cells that give rise to differentiated cell lineages: keratinocytes, hairs, as well as sebocytes. Till now the molecular mechanisms responsible for stem cell quiescent, proliferation, and differentiation have not been fully established. It is suggested that epidermal stem cells might change their fate, both due to intrinsic events and as a result of niche-dependent extrinsic signals; however other yet unknown factors may also be involved in this process. Given the increasing excitement evoked by self-renewing epidermal stem cells, as one of the sources of adult stem cells, it seems important to reveal the mechanisms that govern their fate. In this chapter, we describe recent advances in the characterisation of the epidermal stem cells and their compartments. Furthermore, we focus on the interplay between epidermal stem cells and extrinsic signals and their role in quiescence, proliferation, and differentiation of appropriate epidermal stem cell lineages.
Collapse
|
132
|
Helling AL, Viswanathan P, Cheliotis KS, Mobasseri SA, Yang Y, El Haj AJ, Watt FM. Dynamic Culture Substrates That Mimic the Topography of the Epidermal-Dermal Junction. Tissue Eng Part A 2018; 25:214-223. [PMID: 30280972 PMCID: PMC6388717 DOI: 10.1089/ten.tea.2018.0125] [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] [Indexed: 01/09/2023] Open
Abstract
IMPACT STATEMENT In human skin the junction between the epidermis and dermis undulates. Epidermal stem cells pattern according to their position relative to those undulations. Here we describe a rig in which epidermal cells are cultured on a collagen-coated poly(d,l-lactide-co-glycolide) (PLGA) membrane. When a vacuum is applied the membrane is induced to undulate. Stem cells cluster in response to the vacuum, whereas differentiating cells do not. Rho kinase inhibition results in loss of clustering, suggesting a role for Rho family members in the process. This dynamic platform is a new tool for investigating changes in the skin with age and disease.
Collapse
Affiliation(s)
- Ayelen L Helling
- 1 Center for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Priyalakshmi Viswanathan
- 1 Center for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Katerina S Cheliotis
- 1 Center for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Seyedeh Atefeh Mobasseri
- 1 Center for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Ying Yang
- 2 Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, United Kingdom
| | - Alicia J El Haj
- 2 Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, United Kingdom
| | - Fiona M Watt
- 1 Center for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, United Kingdom
| |
Collapse
|
133
|
Andreeva ER, Matveeva DK. Multipotent Mesenchymal Stromal Cells and Extracellular Matrix: Regulation under Hypoxia. ACTA ACUST UNITED AC 2018. [DOI: 10.1134/s0362119718060038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
134
|
Kitadate Y, Jörg DJ, Tokue M, Maruyama A, Ichikawa R, Tsuchiya S, Segi-Nishida E, Nakagawa T, Uchida A, Kimura-Yoshida C, Mizuno S, Sugiyama F, Azami T, Ema M, Noda C, Kobayashi S, Matsuo I, Kanai Y, Nagasawa T, Sugimoto Y, Takahashi S, Simons BD, Yoshida S. Competition for Mitogens Regulates Spermatogenic Stem Cell Homeostasis in an Open Niche. Cell Stem Cell 2018; 24:79-92.e6. [PMID: 30581080 PMCID: PMC6327111 DOI: 10.1016/j.stem.2018.11.013] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/30/2018] [Accepted: 11/09/2018] [Indexed: 01/08/2023]
Abstract
In many tissues, homeostasis is maintained by physical contact between stem cells and an anatomically defined niche. However, how stem cell homeostasis is achieved in environments where cells are motile and dispersed among their progeny remains unknown. Using murine spermatogenesis as a model, we find that spermatogenic stem cell density is tightly regulated by the supply of fibroblast growth factors (FGFs) from lymphatic endothelial cells. We propose that stem cell homeostasis is achieved through competition for a limited supply of FGFs. We show that the quantitative dependence of stem cell density on FGF dosage, the biased localization of stem cells toward FGF sources, and stem cell dynamics during regeneration following injury can all be predicted and explained within the framework of a minimal theoretical model based on "mitogen competition." We propose that this model provides a generic and robust mechanism to support stem cell homeostasis in open, or facultative, niche environments.
Collapse
Affiliation(s)
- Yu Kitadate
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan; Department of Basic Biology, School of Life Science, Graduate University for Advanced Studies (Sokendai), 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | - David J Jörg
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Cavendish Laboratory, Department of Physics, University of Cambridge, J.J. Thomson Avenue, Cambridge CB3 0HE, UK
| | - Moe Tokue
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan; Department of Basic Biology, School of Life Science, Graduate University for Advanced Studies (Sokendai), 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | - Ayumi Maruyama
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | - Rie Ichikawa
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | - Soken Tsuchiya
- Department of Pharmaceutical Biochemistry, Kumamoto University Graduate School of Pharmaceutical Sciences, Oe-Honmachi, Kumamoto 862-0973, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| | - Eri Segi-Nishida
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-8585, Japan
| | - Toshinori Nakagawa
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan; Department of Basic Biology, School of Life Science, Graduate University for Advanced Studies (Sokendai), 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | - Aya Uchida
- Department of Veterinary Anatomy, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Chiharu Kimura-Yoshida
- Department of Molecular Embryology, Research Institute, Osaka Women's and Children's Hospital, Osaka Prefectural Hospital Organization 840, Murodo-cho, Izumi, Osaka, 594-1101, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Japan
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Japan
| | - Takuya Azami
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Chiyo Noda
- Division of Environmental Photobiology, National Institute for Basic Biology, Okazaki 444-8585, Japan
| | - Satoru Kobayashi
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
| | - Isao Matsuo
- Department of Molecular Embryology, Research Institute, Osaka Women's and Children's Hospital, Osaka Prefectural Hospital Organization 840, Murodo-cho, Izumi, Osaka, 594-1101, Japan
| | - Yoshiakira Kanai
- Department of Veterinary Anatomy, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences, Graduate School of Medicine, Immunology Frontier Research Center, World Premier International Research Center (WPI), Osaka University, Osaka 565-0871, Japan
| | - Yukihiko Sugimoto
- Department of Pharmaceutical Biochemistry, Kumamoto University Graduate School of Pharmaceutical Sciences, Oe-Honmachi, Kumamoto 862-0973, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Japan; Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Japan
| | - Benjamin D Simons
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Cavendish Laboratory, Department of Physics, University of Cambridge, J.J. Thomson Avenue, Cambridge CB3 0HE, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK.
| | - Shosei Yoshida
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan; Department of Basic Biology, School of Life Science, Graduate University for Advanced Studies (Sokendai), 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan.
| |
Collapse
|
135
|
Yoshida S. Open niche regulation of mouse spermatogenic stem cells. Dev Growth Differ 2018; 60:542-552. [PMID: 30443901 PMCID: PMC11520966 DOI: 10.1111/dgd.12574] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/03/2018] [Accepted: 10/03/2018] [Indexed: 02/06/2023]
Abstract
In mammalian testes, robust stem cell functions ensure the continual production of sperm. In testicular seminiferous tubules, spermatogenic stem cells (SSCs) are highly motile and are interspersed between their differentiating progeny, while undergoing self-renewal and differentiation. In such an "open niche" microenvironment, some SSCs proliferate, while others exit the stem cell compartment through differentiation; therefore, self-renewal and differentiation are perfectly balanced at the population (or tissue) level, a dynamics termed "population asymmetry." This is in stark contrast to the classical perception of tissue stem cells being cells that are clustered in a specialized "closed niche" region and that invariantly undergo asymmetric divisions. However, despite its importance, how the self-renewal and differentiation of SSCs are balanced in an open niche environment is poorly understood. Recent studies have thrown light on the key mechanism that enables SSCs to follow heterogeneous fates, although they are equally exposed to signaling molecules controlling self-renewal and differentiation. In particular, SSCs show heterogeneous susceptibilities to differentiation-promoting signals such as Wnt and retinoic acid. Heterogeneous susceptibility to the ubiquitously distributed fate-controlling extracellular signal might be a key generic mechanism for the heterogeneous fate of tissue stem cells in open niche microenvironments.
Collapse
Affiliation(s)
- Shosei Yoshida
- Division of Germ Cell BiologyNational Institute for Basic BiologyNational Institutes of Natural SciencesOkazakiJapan
- Department of Basic BiologySchool of Life ScienceSOKENDAI (Graduate University for Advanced Studies)OkazakiJapan
| |
Collapse
|
136
|
Patel BB, Sharifi F, Stroud DP, Montazami R, Hashemi NN, Sakaguchi DS. 3D Microfibrous Scaffolds Selectively Promotes Proliferation and Glial Differentiation of Adult Neural Stem Cells: A Platform to Tune Cellular Behavior in Neural Tissue Engineering. Macromol Biosci 2018; 19:e1800236. [DOI: 10.1002/mabi.201800236] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/28/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Bhavika B. Patel
- Department of Genetics Development, and Cell Biology and Neuroscience Program Iowa State University Ames IA 50011 USA
| | - Farrokh Sharifi
- Department of Mechanical Engineering Iowa State University Ames IA 50011 USA
| | - Daniel P. Stroud
- Department of Genetics Development, and Cell Biology, Biology Program Iowa State University Ames IA 50011 USA
| | - Reza Montazami
- Department of Mechanical Engineering Iowa State University Ames IA 50011 USA
| | - Nicole N. Hashemi
- Department of Mechanical Engineering Iowa State University Ames IA 50011 USA
| | - Donald S. Sakaguchi
- Department of Genetics Development, and Cell Biology and Neuroscience Program Iowa State University Ames IA 50011 USA
- Department of Genetics Development, and Cell Biology, Biology Program Iowa State University Ames IA 50011 USA
| |
Collapse
|
137
|
Xiao J, Zhang J, Li X, Dai X, Wang J, He Y, Wei L, Shi J, Gong N. Downregulation of Blimp1 inhibits the maturation of bone marrow-derived dendritic cells. Int J Mol Med 2018; 43:1094-1104. [PMID: 30483767 DOI: 10.3892/ijmm.2018.4000] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 11/21/2018] [Indexed: 11/06/2022] Open
Abstract
Modulation of differentiation of dendritic cells (DCs), which are derived from bone marrow cells, may influence their maturation and consequently regulate their ability to present antigens to alloreactive T lymphocytes. B lymphocyte‑induced maturation protein‑1 (Blimp1) is a master regulator of immunocyte differentiation, which has been investigated for its effect on DCs. In the present study, a lentivirus was used as a vector to transduce Blimp1‑short hairpin (sh)RNA into primary bone marrow cells during their differentiation to DCs. Lentiviral‑mediated Blimp1‑shRNA (lenti‑shRNA‑Blimp1) had a transduction efficiency of >60% in DC precursors. Lenti‑shRNA‑Blimp1 significantly downregulated the expression levels of Blimp1 and modulated the expression of its target proteins, including class II major histocompatibility complex (MHC) transactivator, c‑myc and interleukin‑6. Although lenti‑shRNA‑Blimp1 did not interfere with the differentiation of bone marrow cells to DCs, it inhibited DC maturation by decreasing the expression of surface MHC‑II molecules, but not the expression of MHC‑I molecules and co‑stimulatory molecules [cluster of differentiation (CD)80/CD86]. Subsequently, alloreactive T cell proliferation was alleviated and regulatory T cells were expanded in response to lenti‑shRNA‑Blimp1. A toxicity assay indicated that the morphology and proliferation of cultured DCs were mildly influenced by the lentiviral vector, indicating that the use of alternative vectors with minimal or no toxicity could be investigated in future studies. In conclusion, transduction with lenti‑shRNA‑Blimp1 modulated the maturation of DCs via MHC‑II molecule suppression and inhibited alloreactive T cell activation. The present findings supported the application of Blimp1‑based intervention as a novel approach to induce immature DCs for further immunological research.
Collapse
Affiliation(s)
- Jiansheng Xiao
- Department of Hepatobiliary and Organ Transplantation Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ji Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xing Li
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiaomin Dai
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jing Wang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ying He
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Lai Wei
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jun Shi
- Department of Hepatobiliary and Organ Transplantation Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Nianqiao Gong
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
138
|
Barui A, Datta P. Biophysical factors in the regulation of asymmetric division of stem cells. Biol Rev Camb Philos Soc 2018; 94:810-827. [PMID: 30467934 DOI: 10.1111/brv.12479] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/14/2018] [Accepted: 10/18/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Ananya Barui
- Centre for Healthcare Science and TechnologyIndian Institute of Engineering Science and Technology, Shibpur Howrah West Bengal 711103 India
| | - Pallab Datta
- Centre for Healthcare Science and TechnologyIndian Institute of Engineering Science and Technology, Shibpur Howrah West Bengal 711103 India
| |
Collapse
|
139
|
Hosseini FS, Soleimanifar F, Khojasteh A, Ardeshirylajimi A. Promoting osteogenic differentiation of human‐induced pluripotent stem cells by releasing Wnt/β‐catenin signaling activator from the nanofibers. J Cell Biochem 2018; 120:6339-6346. [DOI: 10.1002/jcb.27921] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/25/2018] [Indexed: 01/27/2023]
Affiliation(s)
- Fatemeh Sadat Hosseini
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Fatemeh Soleimanifar
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences Karaj Iran
| | - Arash Khojasteh
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Abdolreza Ardeshirylajimi
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
| |
Collapse
|
140
|
Elis S, Desmarchais A, Cardona E, Fouchecourt S, Dalbies-Tran R, Nguyen T, Thermes V, Maillard V, Papillier P, Uzbekova S, Bobe J, Couderc JL, Monget P. Genes Involved in Drosophila melanogaster Ovarian Function Are Highly Conserved Throughout Evolution. Genome Biol Evol 2018; 10:2629-2642. [PMID: 30060195 PMCID: PMC6173279 DOI: 10.1093/gbe/evy158] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2018] [Indexed: 12/11/2022] Open
Abstract
This work presents a systematic approach to study the conservation of genes between fruit flies and mammals. We have listed 971 Drosophila genes involved in female reproduction at the ovarian level and systematically looked for orthologs in the Ciona, zebrafish, coelacanth, lizard, chicken, and mouse. Depending on the species, the percentage of these Drosophila genes with at least one ortholog varies between 69% and 78%. In comparison, only 42% of all the Drosophila genes have an ortholog in the mouse genome (P < 0.0001), suggesting a dramatically higher evolutionary conservation of ovarian genes. The 177 Drosophila genes that have no ortholog in mice and other vertebrates correspond to genes that are involved in mechanisms of oogenesis that are specific to the fruit fly or the insects. Among 759 genes with at least one ortholog in the zebrafish, 73 have an expression enriched in the ovary in this species (RNA-seq data). Among 760 genes that have at least one ortholog in the mouse; 76 and 11 orthologs are reported to be preferentially and exclusively expressed in the mouse ovary, respectively (based on the UniGene expressed sequence tag database). Several of them are already known to play a key role in murine oogenesis and/or to be enriched in the mouse/zebrafish oocyte, whereas others have remained unreported. We have investigated, by RNA-seq and real-time quantitative PCR, the exclusive ovarian expression of 10 genes in fish and mammals. Overall, we have found several novel candidates potentially involved in mammalian oogenesis by an evolutionary approach and using the fruit fly as an animal model.
Collapse
Affiliation(s)
- Sebastien Elis
- UMR PRC, CNRS, IFCE, INRA, Université de Tours, Nouzilly, France
| | | | | | | | | | | | | | | | - Pascal Papillier
- UMR PRC, CNRS, IFCE, INRA, Université de Tours, Nouzilly, France
| | | | | | - Jean-Louis Couderc
- GReD Laboratory, Université Clermont Auvergne - CNRS UMR 6293- INSERM U1103, Clermont-Ferrand, France
| | - Philippe Monget
- UMR PRC, CNRS, IFCE, INRA, Université de Tours, Nouzilly, France
| |
Collapse
|
141
|
Hay JJ, Rodrigo-Navarro A, Petaroudi M, Bryksin AV, García AJ, Barker TH, Dalby MJ, Salmeron-Sanchez M. Bacteria-Based Materials for Stem Cell Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1804310. [PMID: 30209838 DOI: 10.1002/adma.201804310] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/11/2018] [Indexed: 06/08/2023]
Abstract
Materials can be engineered to deliver specific biological cues that control stem cell growth and differentiation. However, current materials are still limited for stem cell engineering as stem cells are regulated by a complex biological milieu that requires spatiotemporal control. Here a new approach of using materials that incorporate designed bacteria as units that can be engineered to control human mesenchymal stem cells (hMSCs), in a highly dynamic-temporal manner, is presented. Engineered Lactococcus lactis spontaneously colonizes a variety of material surfaces (e.g., polymers, metals, and ceramics) and is able to maintain growth and induce differentiation of hMSCs in 2D/3D surfaces and hydrogels. Controlled, dynamic, expression of fibronectin fragments supports stem cell growth, whereas inducible-temporal regulation of secreted bone morphogenetic protein-2 drives osteogenesis in an on-demand manner. This approach enables stem cell technologies using material systems that host symbiotic interactions between eukaryotic and prokaryotic cells.
Collapse
Affiliation(s)
- Jake J Hay
- Centre for the Cellular Microenvironment, University of Glasgow, Glasgow, G12 8LT, UK
| | | | - Michaela Petaroudi
- Centre for the Cellular Microenvironment, University of Glasgow, Glasgow, G12 8LT, UK
| | - Anton V Bryksin
- Molecular Evolution Core Facility, Georgia Institute of Technology, 950 Atlantic Dr NW, Atlanta, GA 30332, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA
| | - Thomas H Barker
- Department of Cell Biology, University of Virginia, 415 Lane Road, Charlottesville, Virginia, VA 22904, USA
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, University of Glasgow, Glasgow, G12 8LT, UK
| | | |
Collapse
|
142
|
Ito K, Bonora M, Ito K. Metabolism as master of hematopoietic stem cell fate. Int J Hematol 2018; 109:18-27. [PMID: 30219988 DOI: 10.1007/s12185-018-2534-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 09/10/2018] [Indexed: 12/13/2022]
Abstract
HSCs have a fate choice when they divide; they can self-renew, producing new HSCs, or produce daughter cells that will mature to become committed cells. Technical challenges, however, have long obscured the mechanics of these choices. Advances in flow-sorting have made possible the purification of HSC populations, but available HSC-enriched fractions still include substantial heterogeneity, and single HSCs have proven extremely difficult to track and observe. Advances in single-cell approaches, however, have led to the identification of a highly purified population of hematopoietic stem cells (HSCs) that make a critical contribution to hematopoietic homeostasis through a preference for self-renewing division. Metabolic cues are key regulators of this cell fate choice, and the importance of controlling the population and quality of mitochondria has recently been highlighted to maintain the equilibrium of HSC populations. Leukemic cells also demand tightly regulated metabolism, and shifting the division balance of leukemic cells toward commitment has been considered as a promising therapeutic strategy. A deeper understanding of precisely how specific modes of metabolism control HSC fate is, therefore, of great biological interest, and more importantly will be critical to the development of new therapeutic strategies that target HSC division balance for the treatment of hematological disease.
Collapse
Affiliation(s)
- Kyoko Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Massimo Bonora
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA.
- Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
- Albert Einstein Cancer Center and Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
143
|
Bock KW. From TCDD-mediated toxicity to searches of physiologic AHR functions. Biochem Pharmacol 2018; 155:419-424. [PMID: 30055148 DOI: 10.1016/j.bcp.2018.07.032] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 07/23/2018] [Indexed: 12/18/2022]
Abstract
TCDD-mediated toxicity of human individuals together with animal studies led to identification of the aryl hydrocarbon receptor (AHR). It was characterized as multifunctional ligand-activated transcription factor and environmental sensor. Comparison of human toxic responses and animal models provide hints to physiologic AHR functions including chemical and microbial defense, homeostasis of stem/progenitor cells and modulation of the immune system in barrier organs such as skin and the gastrointestinal tract. Extrapolation from animals to humans is difficult due to marked species differences and dependence of AHR function on the cellular context. Nevertheless, therapeutic possibilities of AHR agonists and antagonists are in development. The AHR remains challenging and fascinating.
Collapse
Affiliation(s)
- Karl Walter Bock
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Wilhelmstrasse 56, D-72074 Tübingen, Germany.
| |
Collapse
|
144
|
Wodarz D. Effect of cellular de-differentiation on the dynamics and evolution of tissue and tumor cells in mathematical models with feedback regulation. J Theor Biol 2018; 448:86-93. [PMID: 29605227 PMCID: PMC6173950 DOI: 10.1016/j.jtbi.2018.03.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 03/26/2018] [Accepted: 03/28/2018] [Indexed: 12/12/2022]
Abstract
Tissues are maintained by adult stem cells that self-renew and also differentiate into functioning tissue cells. Homeostasis is achieved by a set of complex mechanisms that involve regulatory feedback loops. Similarly, tumors are believed to be maintained by a minority population of cancer stem cells, while the bulk of the tumor is made up of more differentiated cells, and there is indication that some of the feedback loops that operate in tissues continue to be functional in tumors. Mathematical models of such tissue hierarchies, including feedback loops, have been analyzed in a variety of different contexts. Apart from stem cells giving rise to differentiated cells, it has also been observed that more differentiated cells can de-differentiate into stem cells, both in healthy tissue and tumors, aspects of which have also been investigated mathematically. This paper analyses the effect of de-differentiation on the basic and evolutionary dynamics of cells in the context of tissue hierarchy models that include negative feedback regulation of the cell populations. The models predict that in the presence of de-differentiation, the fixation probability of a neutral mutant is lower than in its absence. Therefore, if de-differentiation occurs, a mutant with identical parameters compared to the wild-type cell population behaves like a disadvantageous mutant. Similarly, the process of de-differentiation is found to lower the fixation probability of an advantageous mutant. These results indicate that the presence of de-differentiation can lower the rates of tumor initiation and progression in the context of the models considered here.
Collapse
Affiliation(s)
- Dominik Wodarz
- Department of Ecology and Evolutionary Biology & Department of Mathematics, 321 Steinhaus Hall, University of California, Irvine, CA 92617, USA.
| |
Collapse
|
145
|
Facchin F, Bianconi E, Canaider S, Basoli V, Biava PM, Ventura C. Tissue Regeneration without Stem Cell Transplantation: Self-Healing Potential from Ancestral Chemistry and Physical Energies. Stem Cells Int 2018; 2018:7412035. [PMID: 30057626 PMCID: PMC6051063 DOI: 10.1155/2018/7412035] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 06/20/2018] [Indexed: 12/17/2022] Open
Abstract
The human body constantly regenerates after damage due to the self-renewing and differentiating properties of its resident stem cells. To recover the damaged tissues and regenerate functional organs, scientific research in the field of regenerative medicine is firmly trying to understand the molecular mechanisms through which the regenerative potential of stem cells may be unfolded into a clinical application. The finding that some organisms are capable of regenerative processes and the study of conserved evolutionary patterns in tissue regeneration may lead to the identification of natural molecules of ancestral species capable to extend their regenerative potential to human tissues. Such a possibility has also been strongly suggested as a result of the use of physical energies, such as electromagnetic fields and mechanical vibrations in human adult stem cells. Results from scientific studies on stem cell modulation confirm the possibility to afford a chemical manipulation of stem cell fate in vitro and pave the way to the use of natural molecules, as well as electromagnetic fields and mechanical vibrations to target human stem cells in their niche inside the body, enhancing human natural ability for self-healing.
Collapse
Affiliation(s)
- Federica Facchin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB) - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Eva Bianconi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB) - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Silvia Canaider
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB) - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Valentina Basoli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| | - Pier Mario Biava
- Scientific Institute of Research and Care Multimedica, Via Milanese 300, 20099 Sesto San Giovanni, Italy
| | - Carlo Ventura
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB) - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| |
Collapse
|
146
|
Weisenberger MS, Deans TL. Bottom-up approaches in synthetic biology and biomaterials for tissue engineering applications. J Ind Microbiol Biotechnol 2018; 45:599-614. [PMID: 29552703 PMCID: PMC6041164 DOI: 10.1007/s10295-018-2027-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 03/11/2018] [Indexed: 12/30/2022]
Abstract
Synthetic biologists use engineering principles to design and construct genetic circuits for programming cells with novel functions. A bottom-up approach is commonly used to design and construct genetic circuits by piecing together functional modules that are capable of reprogramming cells with novel behavior. While genetic circuits control cell operations through the tight regulation of gene expression, a diverse array of environmental factors within the extracellular space also has a significant impact on cell behavior. This extracellular space offers an addition route for synthetic biologists to apply their engineering principles to program cell-responsive modules within the extracellular space using biomaterials. In this review, we discuss how taking a bottom-up approach to build genetic circuits using DNA modules can be applied to biomaterials for controlling cell behavior from the extracellular milieu. We suggest that, by collectively controlling intrinsic and extrinsic signals in synthetic biology and biomaterials, tissue engineering outcomes can be improved.
Collapse
Affiliation(s)
| | - Tara L Deans
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
147
|
Cianflone E, Aquila I, Scalise M, Marotta P, Torella M, Nadal-Ginard B, Torella D. Molecular basis of functional myogenic specification of Bona Fide multipotent adult cardiac stem cells. Cell Cycle 2018; 17:927-946. [PMID: 29862928 PMCID: PMC6103696 DOI: 10.1080/15384101.2018.1464852] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/01/2018] [Accepted: 04/08/2018] [Indexed: 01/14/2023] Open
Abstract
Ischemic Heart Disease (IHD) remains the developed world's number one killer. The improved survival from Acute Myocardial Infarction (AMI) and the progressive aging of western population brought to an increased incidence of chronic Heart Failure (HF), which assumed epidemic proportions nowadays. Except for heart transplantation, all treatments for HF should be considered palliative because none of the current therapies can reverse myocardial degeneration responsible for HF syndrome. To stop the HF epidemic will ultimately require protocols to reduce the progressive cardiomyocyte (CM) loss and to foster their regeneration. It is now generally accepted that mammalian CMs renew throughout life. However, this endogenous regenerative reservoir is insufficient to repair the extensive damage produced by AMI/IHD while the source and degree of CM turnover remains strongly disputed. Independent groups have convincingly shown that the adult myocardium harbors bona-fide tissue specific cardiac stem cells (CSCs). Unfortunately, recent reports have challenged the identity and the endogenous myogenic capacity of the c-kit expressing CSCs. This has hampered progress and unless this conflict is settled, clinical tests of repair/regenerative protocols are unlikely to provide convincing answers about their clinical potential. Here we review recent data that have eventually clarified the specific phenotypic identity of true multipotent CSCs. These cells when coaxed by embryonic cardiac morphogens undergo a precisely orchestrated myogenic commitment process robustly generating bona-fide functional cardiomyocytes. These data should set the path for the revival of further investigation untangling the regenerative biology of adult CSCs to harness their potential for HF prevention and treatment.
Collapse
Affiliation(s)
- Eleonora Cianflone
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Iolanda Aquila
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Mariangela Scalise
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Pina Marotta
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Michele Torella
- Department of Cardiothoracic Sciences, University of Campania Campus “Salvatore Venuta” Viale Europa- Loc. Germaneto “L. Vanvitelli”, Naples, Italy
| | - Bernardo Nadal-Ginard
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| |
Collapse
|
148
|
Controlling the Interfacial Chemical and Physical Properties for Stem Cell Culture. Top Catal 2018. [DOI: 10.1007/s11244-018-0925-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
149
|
Ito K, Ito K. Hematopoietic stem cell fate through metabolic control. Exp Hematol 2018; 64:1-11. [PMID: 29807063 DOI: 10.1016/j.exphem.2018.05.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/18/2018] [Accepted: 05/19/2018] [Indexed: 01/02/2023]
Abstract
Hematopoietic stem cells maintain a quiescent state in the bone marrow to preserve their self-renewal capacity, but also undergo cell divisions as required. Organelles such as the mitochondria sustain cumulative damage during these cell divisions and this damage may eventually compromise the cells' self-renewal capacity. Hematopoietic stem cell divisions result in either self-renewal or differentiation, with the balance between the two affecting hematopoietic homeostasis directly; however, the heterogeneity of available hematopoietic stem cell-enriched fractions, together with the technical challenges of observing hematopoietic stem cell behavior, has long hindered the analysis of individual hematopoietic stem cells and prevented the elucidation of this process. Recent advances in genetic models, metabolomics analyses, and single-cell approaches have revealed the contributions made to hematopoietic stem cell self-renewal by metabolic cues, mitochondrial biogenesis, and autophagy/mitophagy, which have highlighted mitochondrial quality control as a key factor in the equilibrium of hematopoietic stem cells. A deeper understanding of precisely how specific modes of metabolism control hematopoietic stem cells fate at the single-cell level is therefore not only of great biological interest, but will also have clear clinical implications for the development of therapies for hematological diseases.
Collapse
Affiliation(s)
- Kyoko Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA; Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA; Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Albert Einstein Cancer Center and Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
150
|
Hasan A, Byambaa B, Morshed M, Cheikh MI, Shakoor RA, Mustafy T, Marei HE. Advances in osteobiologic materials for bone substitutes. J Tissue Eng Regen Med 2018; 12:1448-1468. [DOI: 10.1002/term.2677] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 02/04/2018] [Accepted: 04/12/2018] [Indexed: 01/03/2023]
Affiliation(s)
- Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering; Qatar University; Doha Qatar
| | - Batzaya Byambaa
- Center for Biomedical Engineering, Department of Medicine; Brigham and Women's Hospital, Harvard Medical School; Cambridge MA USA
- Harvard-MIT Division of Health Sciences and Technology; Massachusetts Institute of Technology; Cambridge MA USA
| | - Mahboob Morshed
- School of Life Sciences; Independent University, Bangladesh (IUB); Dhaka Bangladesh
| | - Mohammad Ibrahim Cheikh
- Department of Mechanical Engineering, Faculty of Engineering and Architecture; American University of Beirut; Beirut Lebanon
| | | | - Tanvir Mustafy
- Department of Mechanical Engineering; Ecole Polytechnique de Montreal; Quebec Canada
| | - Hany E. Marei
- Biomedical Research Center; Qatar University; Doha Qatar
| |
Collapse
|