401
|
Sugimori M, Hayakawa Y, Boman BM, Fields JZ, Awaji M, Kozano H, Tamura R, Yamamoto S, Ogata T, Yamada M, Endo S, Kurimoto M, Kuroda S. Discovery of Power-Law Growth in the Self-Renewal of Heterogeneous Glioma Stem Cell Populations. PLoS One 2015; 10:e0135760. [PMID: 26284929 PMCID: PMC4540573 DOI: 10.1371/journal.pone.0135760] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 07/24/2015] [Indexed: 01/06/2023] Open
Abstract
Background Accumulating evidence indicates that cancer stem cells (CSCs) drive tumorigenesis. This suggests that CSCs should make ideal therapeutic targets. However, because CSC populations in tumors appear heterogeneous, it remains unclear how CSCs might be effectively targeted. To investigate the mechanisms by which CSC populations maintain heterogeneity during self-renewal, we established a glioma sphere (GS) forming model, to generate a population in which glioma stem cells (GSCs) become enriched. We hypothesized, based on the clonal evolution concept, that with each passage in culture, heterogeneous clonal sublines of GSs are generated that progressively show increased proliferative ability. Methodology/Principal Findings To test this hypothesis, we determined whether, with each passage, glioma neurosphere culture generated from four different glioma cell lines become progressively proliferative (i.e., enriched in large spheres). Rather than monitoring self-renewal, we measured heterogeneity based on neurosphere clone sizes (#cells/clone). Log-log plots of distributions of clone sizes yielded a good fit (r>0.90) to a straight line (log(% total clones) = k*log(#cells/clone)) indicating that the system follows a power-law (y = xk) with a specific degree exponent (k = −1.42). Repeated passaging of the total GS population showed that the same power-law was maintained over six passages (CV = −1.01 to −1.17). Surprisingly, passage of either isolated small or large subclones generated fully heterogeneous populations that retained the original power-law-dependent heterogeneity. The anti-GSC agent Temozolomide, which is well known as a standard therapy for glioblastoma multiforme (GBM), suppressed the self-renewal of clones, but it never disrupted the power-law behavior of a GS population. Conclusions/Significance Although the data above did not support the stated hypothesis, they did strongly suggest a novel mechanism that underlies CSC heterogeneity. They indicate that power-law growth governs the self-renewal of heterogeneous glioma stem cell populations. That the data always fit a power-law suggests that: (i) clone sizes follow continuous, non-random, and scale-free hierarchy; (ii) precise biologic rules that reflect self-organizing emergent behaviors govern the generation of neurospheres. That the power-law behavior and the original GS heterogeneity are maintained over multiple passages indicates that these rules are invariant. These self-organizing mechanisms very likely underlie tumor heterogeneity during tumor growth. Discovery of this power-law behavior provides a mechanism that could be targeted in the development of new, more effective, anti-cancer agents.
Collapse
Affiliation(s)
- Michiya Sugimori
- Department of Integrative Neuroscience, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
- * E-mail:
| | - Yumiko Hayakawa
- Department of Neurosurgery, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| | - Bruce M. Boman
- Center for Translational Cancer Research, Departments of Biology and Mathematics, University of Delaware, Helen F Graham Cancer Center and Research Institute, Newark, DE 19711 United States of America
| | - Jeremy Z. Fields
- Biotechnical Research, CATX, Inc., Gladwyne, PA 19035 United States of America
| | - Miharu Awaji
- Department of Integrative Neuroscience, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| | - Hiroko Kozano
- Department of Integrative Neuroscience, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| | - Ryoi Tamura
- Department of Integrative Neuroscience, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| | - Seiji Yamamoto
- Department of Pathology, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| | - Toru Ogata
- The Center of Sports Science and Health Promotion in the NRCD Hospital, National Rehabilitation Center for Persons with Disabilities, 4–1 Namiki, Tokorozawa, Saitama 359–8555, Japan
| | - Mitsuhiko Yamada
- Department of Neuropsycopharmacology, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187–8553, Japan
| | - Shunro Endo
- Department of Neurosurgery, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| | - Masanori Kurimoto
- Department of Neurosurgery, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| | - Satoshi Kuroda
- Department of Neurosurgery, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| |
Collapse
|
402
|
Andreu V, Mendoza G, Arruebo M, Irusta S. Smart Dressings Based on Nanostructured Fibers Containing Natural Origin Antimicrobial, Anti-Inflammatory, and Regenerative Compounds. MATERIALS (BASEL, SWITZERLAND) 2015; 8:5154-5193. [PMID: 28793497 PMCID: PMC5455515 DOI: 10.3390/ma8085154] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 07/30/2015] [Accepted: 08/05/2015] [Indexed: 02/07/2023]
Abstract
A fast and effective wound healing process would substantially decrease medical costs, wound care supplies, and hospitalization significantly improving the patients' quality of life. The search for effective therapeutic approaches seems to be imperative in order to avoid the aggravation of chronic wounds. In spite of all the efforts that have been made during the recent years towards the development of artificial wound dressings, none of the currently available options combine all the requirements necessary for quick and optimal cutaneous regeneration. Therefore, technological advances in the area of temporary and permanent smart dressings for wound care are required. The development of nanoscience and nanotechnology can improve the materials and designs used in topical wound care in order to efficiently release antimicrobial, anti-inflammatory and regenerative compounds speeding up the endogenous healing process. Nanostructured dressings can overcome the limitations of the current coverings and, separately, natural origin components can also overcome the drawbacks of current antibiotics and antiseptics (mainly cytotoxicity, antibiotic resistance, and allergies). The combination of natural origin components with demonstrated antibiotic, regenerative, or anti-inflammatory properties together with nanostructured materials is a promising approach to fulfil all the requirements needed for the next generation of bioactive wound dressings. Microbially compromised wounds have been treated with different essential oils, honey, cationic peptides, aloe vera, plant extracts, and other natural origin occurring antimicrobial, anti-inflammatory, and regenerative components but the available evidence is limited and insufficient to be able to draw reliable conclusions and to extrapolate those findings to the clinical practice. The evidence and some promising preliminary results indicate that future comparative studies are justified but instead of talking about the beneficial or inert effects of those natural origin occurring materials, the scientific community leads towards the identification of the main active components involved and their mechanism of action during the corresponding healing, antimicrobial, or regenerative processes and in carrying out systematic and comparative controlled tests. Once those natural origin components have been identified and their efficacy validated through solid clinical trials, their combination within nanostructured dressings can open up new avenues in the fabrication of bioactive dressings with outstanding characteristics for wound care. The motivation of this work is to analyze the state of the art in the use of different essential oils, honey, cationic peptides, aloe vera, plant extracts, and other natural origin occurring materials as antimicrobial, anti-inflammatory and regenerative components with the aim of clarifying their potential clinical use in bioactive dressings. We conclude that, for those natural occurring materials, more clinical trials are needed to reach a sufficient level of evidence as therapeutic agents for wound healing management.
Collapse
Affiliation(s)
- Vanesa Andreu
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Mariano Esquillor S/N, 50018 Zaragoza, Spain.
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid 28029, Spain.
| | - Gracia Mendoza
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Mariano Esquillor S/N, 50018 Zaragoza, Spain.
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid 28029, Spain.
| | - Manuel Arruebo
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Mariano Esquillor S/N, 50018 Zaragoza, Spain.
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid 28029, Spain.
| | - Silvia Irusta
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Mariano Esquillor S/N, 50018 Zaragoza, Spain.
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid 28029, Spain.
| |
Collapse
|
403
|
Park JK, Yang W, Katsnelson J, Lavker RM, Peng H. MicroRNAs Enhance Keratinocyte Proliferative Capacity in a Stem Cell-Enriched Epithelium. PLoS One 2015; 10:e0134853. [PMID: 26248284 PMCID: PMC4527697 DOI: 10.1371/journal.pone.0134853] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/14/2015] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs are critical regulators of stem cell behavior. The miR-103/107 family is preferentially expressed in the stem cell-enriched corneal limbal epithelium and plays an important role in coordinating several intrinsic characteristics of limbal epithelial stem cells. To elucidate further the mechanisms by which miRs-103/107 function in regulating limbal epithelial stem cells, we investigate the global effects of miRs-103/107 on gene expression in an unbiased manner. Using antagomirs-103/107, we knocked down endogenous miRs-103/107 in keratinocytes and conducted an mRNA profiling study. We show that miRs-103/107 target mitogen-activated protein kinase kinase kinase 7 (MAP3K7) and thereby negatively regulate the p38/AP-1 pathway, which directs epithelial cells towards a differentiated state. Pharmacological inhibition of p38 increases holoclone colony formation, a measure of proliferative capacity. This suggests that the negative regulation of p38 by miRs-103/107 contributes to enhanced proliferative capacity, which is a hallmark of stem cells. Since miRs-103/107 also promote increased holoclone colony formation by regulating JNK activation through non-canonical Wnt signaling, we believe that this microRNA family preserves “stemness” by mediating the crosstalk between the Wnt/JNK and MAP3K7/p38/AP-1 pathways.
Collapse
Affiliation(s)
- Jong Kook Park
- Department of Dermatology, Northwestern University, Chicago, Illinois, United States of America
| | - Wending Yang
- Department of Dermatology, Northwestern University, Chicago, Illinois, United States of America
| | - Julia Katsnelson
- Rush University Medical Center, Chicago, Illinois, United States of America
| | - Robert M. Lavker
- Department of Dermatology, Northwestern University, Chicago, Illinois, United States of America
| | - Han Peng
- Department of Dermatology, Northwestern University, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
404
|
Zhang H, Zhang S, Zhao H, Qiao J, Liu S, Deng Z, Lei X, Ning L, Cao Y, Zhao Y, Duan E. Ovine Hair Follicle Stem Cells Derived from Single Vibrissae Reconstitute Haired Skin. Int J Mol Sci 2015; 16:17779-97. [PMID: 26247934 PMCID: PMC4581221 DOI: 10.3390/ijms160817779] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 07/27/2015] [Accepted: 07/28/2015] [Indexed: 12/17/2022] Open
Abstract
Hair follicle stem cells (HFSCs) possess fascinating self-renewal capacity and multipotency, which play important roles in mammalian hair growth and skin wound repair. Although HFSCs from other mammalian species have been obtained, the characteristics of ovine HFSCs, as well as the methods to isolate them have not been well addressed. Here, we report an efficient strategy to obtain multipotent ovine HFSCs. Through microdissection and organ culture, we obtained keratinocytes that grew from the bulge area of vibrissa hair follicles, and even abundant keratinocytes were harvested from a single hair follicle. These bulge-derived keratinocytes are highly positive for Krt15, Krt14, Tp63, Krt19 and Itga6; in addition to their strong proliferation abilities in vitro, these keratinocytes formed new epidermis, hair follicles and sebaceous glands in skin reconstitution experiments, showing that these are HFSCs from the bulge outer root sheath. Taken together, we developed an efficient in vitro system to enrich ovine HFSCs, providing enough HFSCs for the investigations about the ovine hair cycle, aiming to promote wool production in the future.
Collapse
Affiliation(s)
- Huishan Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Shoubing Zhang
- Department of Histology & Embryology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Huashan Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Jingqiao Qiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Shuang Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Zhili Deng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Xiaohua Lei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Lina Ning
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Yujing Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Enkui Duan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
405
|
Pastushenko I, Prieto-Torres L, Gilaberte Y, Blanpain C. Skin Stem Cells: At the Frontier Between the Laboratory and Clinical Practice. Part 1: Epidermal Stem Cells. ACTAS DERMO-SIFILIOGRAFICAS 2015; 106:725-32. [PMID: 26189363 DOI: 10.1016/j.ad.2015.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 05/26/2015] [Accepted: 05/27/2015] [Indexed: 11/30/2022] Open
Abstract
Stem cells are characterized by their ability to self-renew and differentiate into the different cell lineages of their tissue of origin. The discovery of stem cells in adult tissues, together with the description of specific markers for their isolation, has opened up new lines of investigation, expanding the horizons of biomedical research and raising new hope in the treatment of many diseases. In this article, we review in detail the main characteristics of the stem cells that produce the specialized cells of the skin (epidermal, mesenchymal, and melanocyte stem cells) and their potential implications and applications in diseases affecting the skin. Part I deals with the principal characteristics and potential applications of epidermal stem cells in dermatology.
Collapse
Affiliation(s)
- I Pastushenko
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Bruselas, Bélgica.
| | - L Prieto-Torres
- Servicio de Dermatología, Hospital Clínico Lozano Blesa, Zaragoza, España
| | - Y Gilaberte
- Servicio de Dermatología, Hospital San Jorge, Huesca, España; Instituto Aragonés de Ciencias de la Salud, Zaragoza, España
| | - C Blanpain
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Bruselas, Bélgica; Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Université Libre de Bruxelles (ULB), Bruselas, Bélgica
| |
Collapse
|
406
|
Korem Y, Szekely P, Hart Y, Sheftel H, Hausser J, Mayo A, Rothenberg ME, Kalisky T, Alon U. Geometry of the Gene Expression Space of Individual Cells. PLoS Comput Biol 2015; 11:e1004224. [PMID: 26161936 PMCID: PMC4498931 DOI: 10.1371/journal.pcbi.1004224] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 03/04/2015] [Indexed: 12/14/2022] Open
Abstract
There is a revolution in the ability to analyze gene expression of single cells in a tissue. To understand this data we must comprehend how cells are distributed in a high-dimensional gene expression space. One open question is whether cell types form discrete clusters or whether gene expression forms a continuum of states. If such a continuum exists, what is its geometry? Recent theory on evolutionary trade-offs suggests that cells that need to perform multiple tasks are arranged in a polygon or polyhedron (line, triangle, tetrahedron and so on, generally called polytopes) in gene expression space, whose vertices are the expression profiles optimal for each task. Here, we analyze single-cell data from human and mouse tissues profiled using a variety of single-cell technologies. We fit the data to shapes with different numbers of vertices, compute their statistical significance, and infer their tasks. We find cases in which single cells fill out a continuum of expression states within a polyhedron. This occurs in intestinal progenitor cells, which fill out a tetrahedron in gene expression space. The four vertices of this tetrahedron are each enriched with genes for a specific task related to stemness and early differentiation. A polyhedral continuum of states is also found in spleen dendritic cells, known to perform multiple immune tasks: cells fill out a tetrahedron whose vertices correspond to key tasks related to maturation, pathogen sensing and communication with lymphocytes. A mixture of continuum-like distributions and discrete clusters is found in other cell types, including bone marrow and differentiated intestinal crypt cells. This approach can be used to understand the geometry and biological tasks of a wide range of single-cell datasets. The present results suggest that the concept of cell type may be expanded. In addition to discreet clusters in gene-expression space, we suggest a new possibility: a continuum of states within a polyhedron, in which the vertices represent specialists at key tasks. In the past, biological experiments usually pooled together millions of cells, masking the differences between individual cells. Current technology takes a big step forward by measuring gene expression from individual cells. Interpreting this data is challenging because we need to understand how cells are arranged in a high dimensional gene expression space. Here we test recent theory that suggests that cells facing multiple tasks should be arranged in simple low dimensional polygons or polyhedra (generally called polytopes). The vertices of the polytopes are gene expression profiles optimal for each of the tasks. We find evidence for such simplicity in a variety of tissues—spleen, bone marrow, intestine—analyzed by different single-cell technologies. We find that cells are distributed inside polytopes, such as tetrahedrons or four-dimensional simplexes, with cells closest to each vertex responsible for a different key task. For example, intestinal progenitor cells that give rise to the other cell types show a continuous distribution in a tetrahedron whose vertices correspond to several key sub-tasks. Immune dendritic cells likewise are continuously distributed between key immune tasks. This approach of testing whether data falls in polytopes may be useful for interpreting a variety of single-cell datasets in terms of biological tasks.
Collapse
Affiliation(s)
- Yael Korem
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Pablo Szekely
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yuval Hart
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hila Sheftel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jean Hausser
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Avi Mayo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Michael E. Rothenberg
- Department of Medicine, Division of Gastroenterology and Hepatology, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, United States of America
| | - Tomer Kalisky
- Faculty of Engineering, Bar Ilan University, Ramat Gan, Israel
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|
407
|
Dyment NA, Galloway JL. Regenerative biology of tendon: mechanisms for renewal and repair. ACTA ACUST UNITED AC 2015; 1:124-131. [PMID: 26389023 DOI: 10.1007/s40610-015-0021-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Understanding the molecular and cellular mechanisms underlying tissue turnover and repair are essential towards addressing pathologies in aging, injury and disease. Each tissue has distinct means of maintaining homeostasis and healing after injury. For some, resident stem cell populations mediate both of these processes. These stem cells, by definition, are self renewing and give rise to all the differentiated cells of that tissue. However, not all organs fit with this traditional stem cell model of regeneration, and some do not appear to harbor somatic stem or progenitor cells capable of multilineage in vivo reconstitution. Despite recent progress in tendon progenitor cell research, our current knowledge of the mechanisms regulating tendon cell homeostasis and injury response is limited. Understanding the role of resident tendon cell populations is of great importance for regenerative medicine based approaches to tendon injuries and disease. The goal of this review is to bring to light our current knowledge regarding tendon progenitor cells and their role in tissue maintenance and repair. We will focus on pressing questions in the field and the new tools, including model systems, available to address them.
Collapse
Affiliation(s)
- Nathaniel A Dyment
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center
| | - Jenna L Galloway
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Harvard Stem Cell Institute
| |
Collapse
|
408
|
Pinto-Teixeira F, Viader-Llargués O, Torres-Mejía E, Turan M, González-Gualda E, Pola-Morell L, López-Schier H. Inexhaustible hair-cell regeneration in young and aged zebrafish. Biol Open 2015; 4:903-9. [PMID: 26002931 PMCID: PMC4571094 DOI: 10.1242/bio.012112] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Animals have evolved two general strategies to counter injury and maintain physiological function. The most prevalent is protection by isolating vital organs into body cavities. However, protection is not optimal for sensory systems because their external components need to be exposed to the environment to fulfill their receptive function. Thus, a common strategy to maintain sensory abilities against persistent environmental insult involves repair and regeneration. However, whether age or frequent injuries affect the regenerative capacity of sensory organs remains unknown. We have found that neuromasts of the zebrafish lateral line regenerate mechanosensory hair cells after recurrent severe injuries and in adulthood. Moreover, neuromasts can reverse transient imbalances of Notch signaling that result in defective organ proportions during repair. Our results reveal inextinguishable hair-cell regeneration in the lateral line, and suggest that the neuromast epithelium is formed by plastic territories that are maintained by continuous intercellular communication.
Collapse
Affiliation(s)
- Filipe Pinto-Teixeira
- Laboratory of Sensory Cell Biology & Organogenesis, Centre for Genomic Regulation, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Oriol Viader-Llargués
- Laboratory of Sensory Cell Biology & Organogenesis, Centre for Genomic Regulation, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Elen Torres-Mejía
- Laboratory of Sensory Cell Biology & Organogenesis, Centre for Genomic Regulation, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Melissa Turan
- Laboratory of Sensory Cell Biology & Organogenesis, Centre for Genomic Regulation, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Estela González-Gualda
- Laboratory of Sensory Cell Biology & Organogenesis, Centre for Genomic Regulation, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Laura Pola-Morell
- Laboratory of Sensory Cell Biology & Organogenesis, Centre for Genomic Regulation, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Hernán López-Schier
- Laboratory of Sensory Cell Biology & Organogenesis, Centre for Genomic Regulation, Dr. Aiguader 88, 08003 Barcelona, Spain
| |
Collapse
|
409
|
Abstract
The most common diseases of the joints and its tissues are osteoarthritis and rheumatoid arthritis, with osteoarthritis being anticipated to be the fourth leading cause of disability by the year 2020. To date, no truly causal therapies are available, and this has promoted tissue engineering attempts mainly involving mesenchymal stem cells. The goal of all tissue repairs would be to restore a fully functional tissue, here a hyaline articular cartilage. The hyaline cartilage is the most affected in osteoarthritis, where altered cell–matrix interactions gradually destroy tissue integrity. In rheumatoid arthritis, the inflammatory aspect is more important, and the cartilage tissue is destroyed by the invasion of tumor-like pannus tissue arising from the inflamed synovia. Furthermore, the fibrocartilage of the meniscus is clearly involved in the initiation of osteoarthritis, especially after trauma. Recent investigations have highlighted the role of migratory progenitor cells found in diseased tissues in situ. In osteoarthritis and rheumatoid arthritis, these chondrogenic progenitor cells are involved in regeneration efforts that are largely unsuccessful in diseased cartilage tissue. However, these progenitor cells are interesting targets for a cell-based regenerative therapy for joint diseases.
Collapse
Affiliation(s)
- Boris Schminke
- Tissue Regeneration Work Group, Department of Prosthodontics, Georg August University, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | | |
Collapse
|
410
|
Thompson CM, Wolf JC, Elbekai RH, Paranjpe MG, Seiter JM, Chappell MA, Tappero RV, Suh M, Proctor DM, Bichteler A, Haws LC, Harris MA. Duodenal crypt health following exposure to Cr(VI): Micronucleus scoring, γ-H2AX immunostaining, and synchrotron X-ray fluorescence microscopy. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2015; 789-790:61-6. [PMID: 26232259 DOI: 10.1016/j.mrgentox.2015.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 05/07/2015] [Accepted: 05/10/2015] [Indexed: 12/18/2022]
Abstract
Lifetime exposure to high concentrations of hexavalent chromium [Cr(VI)] in drinking water results in intestinal damage and an increase in duodenal tumors in B6C3F1 mice. To assess whether these tumors could be the result of a direct mutagenic or genotoxic mode of action, we conducted a GLP-compliant 7-day drinking water study to assess crypt health along the entire length of the duodenum. Mice were exposed to water (vehicle control), 1.4, 21, or 180 ppm Cr(VI) via drinking water for 7 consecutive days. Crypt enterocytes in Swiss roll sections were scored as normal, mitotic, apoptotic, karyorrhectic, or as having micronuclei. A single oral gavage of 50mg/kg cyclophosphamide served as a positive control for micronucleus induction. Exposure to 21 and 180 ppm Cr(VI) significantly increased the number of crypt enterocytes. Micronuclei and γ-H2AX immunostaining were not elevated in the crypts of Cr(VI)-treated mice. In contrast, treatment with cyclophosphamide significantly increased numbers of crypt micronuclei and qualitatively increased γ-H2AX immunostaining. Synchrotron-based X-ray fluorescence (XRF) microscopy revealed the presence of strong Cr fluorescence in duodenal villi, but negligible Cr fluorescence in the crypt compartment. Together, these data indicate that Cr(VI) does not adversely effect the crypt compartment where intestinal stem cells reside, and provide additional evidence that the mode of action for Cr(VI)-induced intestinal cancer in B6C3F1 mice involves chronic villous wounding resulting in compensatory crypt enterocyte hyperplasia.
Collapse
Affiliation(s)
| | - Jeffrey C Wolf
- Experimental Pathology Laboratories, Sterling, VA 20166, USA.
| | | | | | - Jennifer M Seiter
- U.S. Army Engineer Research and Development Center, Vicksburg, MS 39180, USA.
| | - Mark A Chappell
- U.S. Army Engineer Research and Development Center, Vicksburg, MS 39180, USA.
| | - Ryan V Tappero
- Photon Sciences Department, Brookhaven National Laboratory, Upton, NY 11973, USA.
| | - Mina Suh
- ToxStrategies, Inc., Mission Viejo, CA 92692, USA.
| | | | | | | | | |
Collapse
|
411
|
Hilmarsdóttir B, Briem E, Sigurdsson V, Franzdóttir SR, Ringnér M, Arason AJ, Bergthorsson JT, Magnusson MK, Gudjonsson T. MicroRNA-200c-141 and ∆Np63 are required for breast epithelial differentiation and branching morphogenesis. Dev Biol 2015; 403:150-61. [PMID: 25967125 DOI: 10.1016/j.ydbio.2015.05.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 04/18/2015] [Accepted: 05/05/2015] [Indexed: 02/06/2023]
Abstract
The epithelial compartment of the breast contains two lineages, the luminal- and the myoepithelial cells. D492 is a breast epithelial cell line with stem cell properties that forms branching epithelial structures in 3D culture with both luminal- and myoepithelial differentiation. We have recently shown that D492 undergo epithelial to mesenchymal transition (EMT) when co-cultured with endothelial cells. This 3D co-culture model allows critical analysis of breast epithelial lineage development and EMT. In this study, we compared the microRNA (miR) expression profiles for D492 and its mesenchymal-derivative D492M. Suppression of the miR-200 family in D492M was among the most profound changes observed. Exogenous expression of miR-200c-141 in D492M reversed the EMT phenotype resulting in gain of luminal but not myoepithelial differentiation. In contrast, forced expression of ∆Np63 in D492M restored the myoepithelial phenotype only. Co-expression of miR-200c-141 and ∆Np63 in D492M restored the branching morphogenesis in 3D culture underlining the requirement for both luminal and myoepithelial elements for obtaining full branching morphogenesis in breast epithelium. Introduction of a miR-200c-141 construct in both D492 and D492M resulted in resistance to endothelial induced EMT. In conclusion, our data suggests that expression of miR-200c-141 and ∆Np63 in D492M can reverse EMT resulting in luminal- and myoepithelial differentiation, respectively, demonstrating the importance of these molecules in epithelial integrity in the human breast.
Collapse
Affiliation(s)
- Bylgja Hilmarsdóttir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland
| | - Eirikur Briem
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland
| | - Valgardur Sigurdsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Sigrídur Rut Franzdóttir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Markus Ringnér
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Sweden
| | - Ari Jon Arason
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland
| | - Jon Thor Bergthorsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland
| | - Magnus Karl Magnusson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland; Department of Medical Pharmacology and Toxicology, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland
| | - Thorarinn Gudjonsson
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Iceland; Department of Laboratory Hematology, Landspitali-University Hospital, Iceland.
| |
Collapse
|
412
|
Wang B, Tan L, Deng D, Lu T, Zhou C, Li Z, Tang Z, Wu Z, Tang H. Novel stable cytokine delivery system in physiological pH solution: chitosan oligosaccharide/heparin nanoparticles. Int J Nanomedicine 2015; 10:3417-27. [PMID: 26056441 PMCID: PMC4431508 DOI: 10.2147/ijn.s82091] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Cell therapy is a promising strategy for tissue regeneration. Key to this strategy is mobilization and recruitment of exogenous or autologous stem/progenitor cells by cytokines. However, there is no effective cytokine delivery system available for clinic application, in particular for myocardial regeneration. The aim of this study was to develop a novel cytokine delivery system that is stable in solution at physiological pH. Methods Four groups of self-assembled chitosan oligosaccharide/heparin (CSO/H) nanoparticles were prepared with various volume ratios of chitosan oligosaccharide to heparin (5:2, 5:4, 4:15, 1:5) and characterized by laser diffraction, particle size analysis, and transmission electron microscopy. The encapsulation efficiency and loading content of two cytokines, ie, stromal cell-derived factor (SDF)-1α and vascular endothelial growth factor (VEGF) were quantified using an enzyme-linked immunosorbent assay. The biological activity of the loaded SDF-1α and VEGF was evaluated using the transwell migration assay and MTT assay. The dispersion profiles for the cytokine-loaded nanoparticles were quantified using fluorescence molecular tomography. Results CSO/H nanoparticles were prepared successfully in solution with physiological pH. The particle sizes in the four treatment groups were in the range of 96.2–210.5 nm and the zeta potential ranged from −29.4 mV to 24.2 mV. The loading efficiency in the CSO/H nanoparticle groups with the first three ratios was more than 90%. SDF-1α loaded into CSO/H nanoparticles retained its migration activity and VEGF loaded into CSO/H nanoparticles continued to show proliferation activity. The in vivo dispersion test showed that the CSO/H nanoparticles enabled to VEGF to accumulate locally for a longer period of time. Conclusion CSO/H nanoparticles have a high cytokine loading capacity and allow cytokines to maintain their bioactivity for longer, are stable in an environment with physiological pH, and may be a promising cytokine delivery system for tissue regeneration.
Collapse
Affiliation(s)
- Bin Wang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Ling Tan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Dengpu Deng
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Ting Lu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Changwei Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Zhongkui Li
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Zhenjie Tang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Zhongshi Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Hao Tang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| |
Collapse
|
413
|
Jain R, Barkauskas CE, Takeda N, Bowie EJ, Aghajanian H, Wang Q, Padmanabhan A, Manderfield LJ, Gupta M, Li D, Li L, Trivedi CM, Hogan BLM, Epstein JA. Plasticity of Hopx(+) type I alveolar cells to regenerate type II cells in the lung. Nat Commun 2015; 6:6727. [PMID: 25865356 PMCID: PMC4396689 DOI: 10.1038/ncomms7727] [Citation(s) in RCA: 218] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 02/23/2015] [Indexed: 12/22/2022] Open
Abstract
The plasticity of differentiated cells in adult tissues undergoing repair is an area of intense research. Pulmonary alveolar type II cells produce surfactant and function as progenitors in the adult, demonstrating both self-renewal and differentiation into gas exchanging type I cells. In vivo, type I cells are thought to be terminally differentiated and their ability to give rise to alternate lineages has not been reported. Here we show that Hopx becomes restricted to type I cells during development. However, unexpectedly, lineage-labelled Hopx(+) cells both proliferate and generate type II cells during adult alveolar regrowth following partial pneumonectomy. In clonal 3D culture, single Hopx(+) type I cells generate organoids composed of type I and type II cells, a process modulated by TGFβ signalling. These findings demonstrate unanticipated plasticity of type I cells and a bidirectional lineage relationship between distinct differentiated alveolar epithelial cell types in vivo and in single-cell culture.
Collapse
Affiliation(s)
- Rajan Jain
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Christina E Barkauskas
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke Medicine, Durham, North Carolina 27710, USA
| | - Norifumi Takeda
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Emily J Bowie
- Department of Cell Biology, Duke Medicine, Durham, North Carolina 27710, USA
| | - Haig Aghajanian
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Qiaohong Wang
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Arun Padmanabhan
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Lauren J Manderfield
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Mudit Gupta
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Deqiang Li
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Li Li
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke Medicine, Durham, North Carolina 27710, USA
| | - Chinmay M Trivedi
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Brigid L M Hogan
- Department of Cell Biology, Duke Medicine, Durham, North Carolina 27710, USA
| | - Jonathan A Epstein
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
414
|
Saito MT, Silvério KG, Casati MZ, Sallum EA, Jr FHN. Tooth-derived stem cells: Update and perspectives. World J Stem Cells 2015; 7:399-407. [PMID: 25815123 PMCID: PMC4369495 DOI: 10.4252/wjsc.v7.i2.399] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 10/22/2014] [Accepted: 11/10/2014] [Indexed: 02/06/2023] Open
Abstract
Tissue engineering is an emerging field of science that focuses on creating suitable conditions for the regeneration of tissues. The basic components for tissue engineering involve an interactive triad of scaffolds, signaling molecules, and cells. In this context, stem cells (SCs) present the characteristics of self-renewal and differentiation capacity, which make them promising candidates for tissue engineering. Although they present some common markers, such as cluster of differentiation (CD)105, CD146 and STRO-1, SCs derived from various tissues have different patterns in relation to proliferation, clonogenicity, and differentiation abilities in vitro and in vivo. Tooth-derived tissues have been proposed as an accessible source to obtain SCs with limited morbidity, and various tooth-derived SCs (TDSCs) have been isolated and characterized, such as dental pulp SCs, SCs from human exfoliated deciduous teeth, periodontal ligament SCs, dental follicle progenitor cells, SCs from apical papilla, and periodontal ligament of deciduous teeth SCs. However, heterogeneity among these populations has been observed, and the best method to select the most appropriate TDSCs for regeneration approaches has not yet been established. The objective of this review is to outline the current knowledge concerning the various types of TDSCs, and discuss the perspectives for their use in regenerative approaches.
Collapse
|
415
|
Adam RC, Yang H, Rockowitz S, Larsen SB, Nikolova M, Oristian DS, Polak L, Kadaja M, Asare A, Zheng D, Fuchs E. Pioneer factors govern super-enhancer dynamics in stem cell plasticity and lineage choice. Nature 2015; 521:366-70. [PMID: 25799994 PMCID: PMC4482136 DOI: 10.1038/nature14289] [Citation(s) in RCA: 303] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 02/06/2015] [Indexed: 12/12/2022]
Abstract
Adult stem cells (SCs) reside in niches which balance self-renewal with lineage selection and progression during tissue homeostasis. Following injury, culture or transplantation, SCs outside their niche often display fate flexibility1-4. Here we show that super-enhancers5 underlie the identity, lineage commitment and plasticity of adult SCs in vivo. Using hair follicle (HF) as model, we map the global chromatin domains of HFSCs and their committed progenitors in their native microenvironments. We show that super-enhancers and their dense clusters (‘epicenters’) of transcription factor (TF) binding sites change upon lineage progression. New fate is acquired by decommissioning old and establishing new super-enhancers and/or epicenters, an auto-regulatory process that abates one master regulator subset while enhancing another. We further show that when outside their niche, either in vitro or in wound-repair, HFSCs dynamically remodel super-enhancers in response to changes in their microenvironment. Intriguingly, some key super-enhancers shift epicenters, enabling them to remain active and maintain a transitional state in an ever-changing transcriptional landscape. Finally, we identify SOX9 as a crucial chromatin rheostat of HFSC super-enhancers, and provide functional evidence that super-enhancers are dynamic, dense TF-binding platforms which are acutely sensitive to pioneer master regulators whose levels define not only spatial and temporal features of lineage-status, but also stemness, plasticity in transitional states and differentiation.
Collapse
Affiliation(s)
- Rene C Adam
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology &Development, The Rockefeller University, New York, New York 10065, USA
| | - Hanseul Yang
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology &Development, The Rockefeller University, New York, New York 10065, USA
| | - Shira Rockowitz
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Samantha B Larsen
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology &Development, The Rockefeller University, New York, New York 10065, USA
| | - Maria Nikolova
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology &Development, The Rockefeller University, New York, New York 10065, USA
| | - Daniel S Oristian
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology &Development, The Rockefeller University, New York, New York 10065, USA
| | - Lisa Polak
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology &Development, The Rockefeller University, New York, New York 10065, USA
| | - Meelis Kadaja
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology &Development, The Rockefeller University, New York, New York 10065, USA
| | - Amma Asare
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology &Development, The Rockefeller University, New York, New York 10065, USA
| | - Deyou Zheng
- 1] Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461, USA [2] Departments of Neurology and Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology &Development, The Rockefeller University, New York, New York 10065, USA
| |
Collapse
|
416
|
Borcherding N, Kusner D, Kolb R, Xie Q, Li W, Yuan F, Velez G, Askeland R, Weigel RJ, Zhang W. Paracrine WNT5A Signaling Inhibits Expansion of Tumor-Initiating Cells. Cancer Res 2015; 75:1972-82. [PMID: 25769722 DOI: 10.1158/0008-5472.can-14-2761] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 03/09/2015] [Indexed: 12/21/2022]
Abstract
It is not well understood how paracrine communication between basal and luminal cell populations in the mammary gland affects tumorigenesis. During ErbB2-induced mammary tumorigenesis, enriched mammary stem cells that represent a subpopulation of basal cells exhibit enhanced tumorigenic capacity compared with the corresponding luminal progenitors. Transcript profiling of tumors derived from basal and luminal tumor-initiating cells (TIC) revealed preferential loss of the noncanonical Wnt ligand WNT5A in basal TIC-derived tumors. Heterozygous loss of WNT5A was correlated with shorter survival of breast cancer patients. In a mouse model of ErbB2-induced breast cancer, Wnt5a heterozygosity promoted tumor multiplicity and pulmonary metastasis. As a TGFβ substrate, luminal cell-produced WNT5A induced a feed-forward loop to activate SMAD2 in a RYK and TGFβR1-dependent manner to limit the expansion of basal TIC in a paracrine fashion, a potential explanation for the suppressive effect of WNT5A in mammary tumorigenesis. Our results identify the WNT5A/RYK module as a spatial regulator of the TGFβ-SMAD signaling pathway in the context of mammary gland development and carcinogenesis, offering a new perspective on tumor suppression provided by basal-luminal cross-talk in normal mammary tissue.
Collapse
Affiliation(s)
- Nicholas Borcherding
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, Iowa. Medical Science Training Program, University of Iowa, College of Medicine, Iowa City, Iowa
| | - David Kusner
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, Iowa. Molecular and Cellular Biology Program, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Ryan Kolb
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, Iowa. Immunology Program, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Qing Xie
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, Iowa. College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, P.R. China
| | - Wei Li
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Fang Yuan
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, Iowa. Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Gabriel Velez
- Medical Science Training Program, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Ryan Askeland
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Ronald J Weigel
- Department of Surgery, University of Iowa, College of Medicine, Iowa City, Iowa. Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Weizhou Zhang
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, Iowa. Medical Science Training Program, University of Iowa, College of Medicine, Iowa City, Iowa. Molecular and Cellular Biology Program, University of Iowa, College of Medicine, Iowa City, Iowa. Immunology Program, University of Iowa, College of Medicine, Iowa City, Iowa. Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, Iowa.
| |
Collapse
|
417
|
Sipos F, Műzes G. Injury-associated reacquiring of intestinal stem cell function. World J Gastroenterol 2015; 21:2005-2010. [PMID: 25717233 PMCID: PMC4326135 DOI: 10.3748/wjg.v21.i7.2005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 11/04/2014] [Accepted: 01/08/2015] [Indexed: 02/06/2023] Open
Abstract
Epithelial layer of the intestine relies upon stem cells for maintaining homeostasis and regeneration. Two types of stem cells are currently defined in intestinal crypts: the cycling crypt base columnar cells and quiescent cells. Though several candidate markers and regulators of rapidly cycling and quiescent stem cells have been identified so far, the exact nature of quiescent cells is still questionable since investigations mainly focused on candidate markers rather than the label-retaining population itself. Recent results, however, have strengthened the argument for functional plasticity. Using a lineage tracing strategy label-retaining cells (LRCs) of the intestinal epithelium were marked, then followed by a pulse-chase analysis it was found that during homeostasis, LRCs were Lgr5-positive and were destined to become Paneth and neuroendocrine cells. Nevertheless, it was demonstrated that LRCs are capable of clonogenic growth by recall to the self-renewing pool of stem cells in case of epithelial injury. These new findings highlight on the hierarchical and spatial organization of intestinal epithelial homeostasis and the important plasticity of progenitors during tissue regeneration, moreover, provide a motivation for studying their role in disorders like colorectal cancer.
Collapse
|
418
|
Crystal RG. Airway basal cells. The "smoking gun" of chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2015; 190:1355-62. [PMID: 25354273 DOI: 10.1164/rccm.201408-1492pp] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The earliest abnormality in the lung associated with smoking is hyperplasia of airway basal cells, the stem/progenitor cells of the ciliated and secretory cells that are central to pulmonary host defense. Using cell biology and 'omics technologies to assess basal cells isolated from bronchoscopic brushings of nonsmokers, smokers, and smokers with chronic obstructive pulmonary disease (COPD), compelling evidence has been provided in support of the concept that airway basal cells are central to the pathogenesis of smoking-associated lung diseases. When confronted by the chronic stress of smoking, airway basal cells become disorderly, regress to a more primitive state, behave as dictated by their inheritance, are susceptible to acquired changes in their genome, lose the capacity to regenerate the epithelium, are responsible for the major changes in the airway that characterize COPD, and, with persistent stress, can undergo malignant transformation. Together, these observations led to the conclusion that accelerated loss of lung function in susceptible individuals begins with disordered airway basal cell biology (i.e., that airway basal cells are the "smoking gun" of COPD, a potential target for the development of therapies to prevent smoking-related lung disorders).
Collapse
Affiliation(s)
- Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
419
|
Hiratsuka T, Fujita Y, Naoki H, Aoki K, Kamioka Y, Matsuda M. Intercellular propagation of extracellular signal-regulated kinase activation revealed by in vivo imaging of mouse skin. eLife 2015; 4:e05178. [PMID: 25668746 PMCID: PMC4337632 DOI: 10.7554/elife.05178] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 01/09/2015] [Indexed: 01/20/2023] Open
Abstract
Extracellular signal-regulated kinase (ERK) is a key effector of many growth signalling pathways. In this study, we visualise epidermal ERK activity in living mice using an ERK FRET biosensor. Under steady-state conditions, the epidermis occasionally revealed bursts of ERK activation patterns where ERK activity radially propagated from cell to cell. The frequency of this spatial propagation of radial ERK activity distribution (SPREAD) correlated with the rate of epidermal cell division. SPREADs and proliferation were stimulated by 12-O-tetradecanoylphorbol 13-acetate (TPA) in a manner dependent on EGF receptors and their cognate ligands. At the wounded skin, ERK activation propagated as trigger wave in parallel to the wound edge, suggesting that ERK activation propagation can be superimposed. Furthermore, by visualising the cell cycle, we found that SPREADs were associated with G2/M cell cycle progression. Our results provide new insights into how cell proliferation and transient ERK activity are synchronised in a living tissue. DOI:http://dx.doi.org/10.7554/eLife.05178.001 Our skin is our largest organ; it provides a barrier that protects the underlying tissues and internal organs from the external environment and acts as one of our first lines of defense against infection. Both of these roles subject the skin to wear and tear and so it must constantly create new skin cells to replace those lost or damaged. However, if this renewal process goes awry it can lead to excessive cell growth or skin cancer. To avoid this, cells tightly regulate the pathways that stimulate skin renewal. Skin renewal involves growth signals activating an enzyme called ERK. When and where the ERK enzyme is activated is normally tightly regulated, and many kinds of cancer have been linked to ERK becoming active at the wrong time or in the wrong place. Despite the importance of ERK in skin cells, a number of technical challenges have made it difficult to study how these signals are passed from cell to cell. Hiratsuka et al. have now examined genetically altered mice that produce a fluorescent sensor molecule that makes it possible to see ERK activity in living skin cells. The skin of anesthetized mice was observed under a microscope, and time-lapse videos revealed occasional ‘firework-like’ bursts of ERK activity. At first the ERK enzyme was active in a small cluster of skin cells, then ERK activity was seen in the surrounding cells—appearing to spread outwards over the course of several minutes—before the activity stopped. Hiratsuka et al. named this pattern of activity a ‘Spatial Propagation of Radial ERK Activity Distribution’, or SPREAD for short. By studying SPREADs in the skin on the ears and the back of these mice, Hiratsuka et al. learned that these bursts of ERK activity coincided with skin cell growth; the bursts happened more frequently in the areas where the skin cells were dividing. Applying a chemical that stimulates cell division to the skin of the mice triggered more bursts of ERK activity; whereas fewer bursts were observed if Hiratsuka et al. used other chemicals to block the activity of some of the signaling proteins that work upstream of ERK. Further experiments suggested that SPREADs encourage cells to progress through the cycle of events that leads a cell to divide; blocking these bursts caused the cell to pause at the stage just before it would normally divide. Hiratsuka et al. also observed similar patterns of ERK activity moving out like waves from the edges of skin wounds. Further research using similar methods will reveal how growth signals are triggered and propagated in healthy and diseased tissues, not only in the skin but also other organs such as the liver, intestine, and muscles. DOI:http://dx.doi.org/10.7554/eLife.05178.002
Collapse
Affiliation(s)
- Toru Hiratsuka
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshihisa Fujita
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Honda Naoki
- Imaging Platform for Spatio-Temporal Information, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuhiro Aoki
- Imaging Platform for Spatio-Temporal Information, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuji Kamioka
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Michiyuki Matsuda
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
420
|
Arnold KM, Opdenaker LM, Flynn D, Sims-Mourtada J. Wound healing and cancer stem cells: inflammation as a driver of treatment resistance in breast cancer. CANCER GROWTH AND METASTASIS 2015; 8:1-13. [PMID: 25674014 PMCID: PMC4315129 DOI: 10.4137/cgm.s11286] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/01/2014] [Accepted: 12/05/2014] [Indexed: 12/13/2022]
Abstract
The relationship between wound healing and cancer has long been recognized. The mechanisms that regulate wound healing have been shown to promote transformation and growth of malignant cells. In addition, chronic inflammation has been associated with malignant transformation in many tissues. Recently, pathways involved in inflammation and wound healing have been reported to enhance cancer stem cell (CSC) populations. These cells, which are highly resistant to current treatments, are capable of repopulating the tumor after treatment, causing local and systemic recurrences. In this review, we highlight proinflammatory cytokines and developmental pathways involved in tissue repair, whose deregulation in the tumor microenvironment may promote growth and survival of CSCs. We propose that the addition of anti-inflammatory agents to current treatment regimens may slow the growth of CSCs and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Kimberly M Arnold
- Center for Translational Cancer Research, Helen F. Graham Cancer Center, Christiana Care Health Services, Inc., Newark, DE, USA. ; Department of Medical Laboratory Sciences, University of Delaware, Newark, DE, USA
| | - Lynn M Opdenaker
- Center for Translational Cancer Research, Helen F. Graham Cancer Center, Christiana Care Health Services, Inc., Newark, DE, USA. ; Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Daniel Flynn
- Center for Translational Cancer Research, Helen F. Graham Cancer Center, Christiana Care Health Services, Inc., Newark, DE, USA. ; Department of Medical Laboratory Sciences, University of Delaware, Newark, DE, USA
| | - Jennifer Sims-Mourtada
- Center for Translational Cancer Research, Helen F. Graham Cancer Center, Christiana Care Health Services, Inc., Newark, DE, USA. ; Department of Medical Laboratory Sciences, University of Delaware, Newark, DE, USA
| |
Collapse
|
421
|
Zhao H, Qiao J, Zhang S, Zhang H, Lei X, Wang X, Deng Z, Ning L, Cao Y, Guo Y, Liu S, Duan E. GPR39 marks specific cells within the sebaceous gland and contributes to skin wound healing. Sci Rep 2015; 5:7913. [PMID: 25604641 PMCID: PMC4300488 DOI: 10.1038/srep07913] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 12/19/2014] [Indexed: 12/28/2022] Open
Abstract
G protein-coupled receptors (GPCRs) mediate multiple key biological processes in the body. The orphan receptor GPR39 has been reported to be involved in various pathophysiological events. However, the function of GPR39 in skin biology remains unknown. Using a genetically engineered mouse strain in which lacZ expression faithfully replaced endogenous Gpr39 expression, we discovered a unique expression pattern of Gpr39 in the sebaceous gland (SG). Using various methods, we confirmed that GPR39 marked a specific cell population at the opening of the SG and colocalised with the SG stem cell marker Blimp1. Further investigations showed that GPR39 was spatiotemporally expressed during skin wound repair. Although it was dispensable for skin development and homeostasis, GPR39 contributed positively to skin wound healing: its loss led to a delay in wound healing during the intermediate stage. The present study reveals a novel role of GPR39 in both dermatology and stem cell biology that has not been previously recognised.
Collapse
Affiliation(s)
- Huashan Zhao
- 1] State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China [2] University of Chinese Academy of Sciences, Beijing, China
| | - Jingqiao Qiao
- College of Animal Science and Technology, Beijing University of Agriculture, China
| | - Shoubing Zhang
- Department of Histology&Embryology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Huishan Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiaohua Lei
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xinyue Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhili Deng
- 1] State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China [2] University of Chinese Academy of Sciences, Beijing, China
| | - Lina Ning
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yujing Cao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yong Guo
- College of Animal Science and Technology, Beijing University of Agriculture, China
| | - Shuang Liu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Enkui Duan
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
422
|
Liu L, Wu L, Wei X, Ling J. Induced overexpression of Oct4A in human dental pulp cells enhances pluripotency and multilineage differentiation capability. Stem Cells Dev 2015; 24:962-72. [PMID: 25422984 DOI: 10.1089/scd.2014.0388] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Octamer-binding transcription factor 4A (Oct4A), one of the three spliced variants of the class V of POU transcription factor family, is mainly expressed in the nucleus of undifferentiated cells and serves as the key regulator for the maintenance of pluripotency and self-renewal. However, its specific role in regulating pluripotency and multilineage differentiation potential of dental pulp cells (DPCs) remains unknown. To explore the effect of Oct4A on pluripotency and multilineage differentiation capability of DPCs, expression of Oct4A in human dental pulp tissue and pluripotent markers Oct4A, Sox2, c-Myc, Nanog, and Klf4 in DPCs with prolonged in vitro culture were examined by immunohistochemistry and immunofluorescent staining. Oct4A transfection rate in DPCs with lentivirus was evaluated by real-time polymerase chain reaction (PCR) and western blot. Cell proliferation, multilineage differentiation, and the expression of Oct4B1, Sox2, Nanog, Klf4, c-Myc, and Utf1 in DPCs after Oct4A transfection were detected by cell counting kit-8, Alizarin red/Oil red O staining, immunofluorescent staining, alkaline phosphatase analysis, and real-time PCR. We demonstrated that Oct4A was mainly expressed in the nucleus of odontoblasts in dental pulp tissue. Oct4A, Sox2, c-Myc, Nanog, and Klf4 were primarily located in the nucleus of DPCs at early passage (passage 1) and translocated to cytoplasm at late passage (passage 7). In DPCs with Oct4A overexpression, Oct4A, Oct4B1, Sox2, Nanog, Klf4, c-Myc, and Utf1 were significantly upregulated (p<0.05) and the cell proliferation (p<0.05), odontogenic and adipogenic differentiation were significantly enhanced. Taken together, Oct4A plays a critical role in regulation of cell proliferation, pluripotency, and multilineage differentiation potential of DPCs.
Collapse
Affiliation(s)
- Lu Liu
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatological Hospital, Guangdong Province Key Laboratory of Stomatology, Sun Yat-sen University , Guangzhou, China
| | | | | | | |
Collapse
|
423
|
Seguin L, Desgrosellier JS, Weis SM, Cheresh DA. Integrins and cancer: regulators of cancer stemness, metastasis, and drug resistance. Trends Cell Biol 2015; 25:234-40. [PMID: 25572304 DOI: 10.1016/j.tcb.2014.12.006] [Citation(s) in RCA: 542] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/03/2014] [Accepted: 12/05/2014] [Indexed: 12/17/2022]
Abstract
Interactions between cancer cells and their surroundings can trigger essential signaling cues that determine cell fate and influence the evolution of the malignant phenotype. As the primary receptors involved in cell-matrix adhesion, integrins present on the surface of tumor and stromal cells have a profound impact on the ability to survive in specific locations, but in some cases, these receptors can also function in the absence of ligand binding to promote stemness and survival in the presence of environmental and therapeutic stresses. Understanding how integrin expression and function is regulated in this context will enable the development of new therapeutic approaches to sensitize tumors to therapy and suppress their metastatic phenotype.
Collapse
Affiliation(s)
- Laetitia Seguin
- Department of Pathology and the Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Jay S Desgrosellier
- Department of Pathology and the Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Sara M Weis
- Department of Pathology and the Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - David A Cheresh
- Department of Pathology and the Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
424
|
Hess S, Rambukkana A. Bacterial-induced cell reprogramming to stem cell-like cells: new premise in host-pathogen interactions. Curr Opin Microbiol 2014; 23:179-88. [PMID: 25541240 DOI: 10.1016/j.mib.2014.11.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 11/26/2014] [Indexed: 12/12/2022]
Abstract
Bacterial pathogens employ a myriad of strategies to alter host tissue cell functions for bacterial advantage during infection. Recent advances revealed a fusion of infection biology with stem cell biology by demonstrating developmental reprogramming of lineage committed host glial cells to progenitor/stem cell-like cells by an intracellular bacterial pathogen Mycobacterium leprae. Acquisition of migratory and immunomodulatory properties of such reprogrammed cells provides an added advantage for promoting bacterial spread. This presents a previously unseen sophistication of cell manipulation by hijacking the genomic plasticity of host cells by a human bacterial pathogen. The rationale for such extreme fate conversion of host cells may be directly linked to the exceedingly passive obligate life style of M. leprae with a degraded genome and host cell dependence for both bacterial survival and dissemination, particularly the use of host-derived stem cell-like cells as a vehicle for spreading infection without being detected by immune cells. Thus, this unexpected link between cell reprogramming and infection opens up a new premise in host-pathogen interactions. Furthermore, such bacterial ingenuity could also be harnessed for developing natural ways of reprogramming host cells for repairing damaged tissues from infection, injury and diseases.
Collapse
Affiliation(s)
- Samuel Hess
- MRC Centre for Regenerative Medicine, University of Edinburgh, Little France Campus, Edinburgh EH16 4UU, United Kingdom
| | - Anura Rambukkana
- MRC Centre for Regenerative Medicine, University of Edinburgh, Little France Campus, Edinburgh EH16 4UU, United Kingdom; Edinburgh Infectious Diseases, University of Edinburgh, Edinburgh, United Kingdom; Centre for Neuroregeneration, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
425
|
Sun BK, Siprashvili Z, Khavari PA. Advances in skin grafting and treatment of cutaneous wounds. Science 2014; 346:941-5. [PMID: 25414301 DOI: 10.1126/science.1253836] [Citation(s) in RCA: 568] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ability of the skin to repair itself after injury is vital to human survival and is disrupted in a spectrum of disorders. The process of cutaneous wound healing is complex, requiring a coordinated response by immune cells, hematopoietic cells, and resident cells of the skin. We review the classic paradigms of wound healing and evaluate how recent discoveries have enriched our understanding of this process. We evaluate current and experimental approaches to treating cutaneous wounds, with an emphasis on cell-based therapies and skin transplantation.
Collapse
Affiliation(s)
- Bryan K Sun
- Program in Epithelial Biology, Stanford University, Stanford, CA 94305, USA
| | - Zurab Siprashvili
- Program in Epithelial Biology, Stanford University, Stanford, CA 94305, USA
| | - Paul A Khavari
- Program in Epithelial Biology, Stanford University, Stanford, CA 94305, USA. Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA 94304, USA.
| |
Collapse
|
426
|
Sonnenschein C, Davis B, Soto AM. A novel pathogenic classification of cancers. Cancer Cell Int 2014; 14:113. [PMID: 25493071 PMCID: PMC4260242 DOI: 10.1186/s12935-014-0113-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/23/2014] [Indexed: 01/28/2023] Open
Abstract
According to contemporary epidemiological and experimental evidence, we propose a novel classification of cancers based on pathogenesis instead of classifications based on histological appearance of cancer. This new scheme first defines cancers as either 1. inborn errors of development or 2. sporadic ones, and then sub-defines the former into 1A. inborn inherited errors of development, being those due to mutations contributed by one or both parents' gametes to the developing conceptus, and 1B. inborn induced errors of development when the malformations and/or cancers are due to environmental carcinogenic exposure during pregnancy. It is anticipated that the origin of an increasing number of so-called sporadic cancers will turn out to be linked to the inborn induced errors of development group.
Collapse
Affiliation(s)
- Carlos Sonnenschein
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Barbara Davis
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Ana M Soto
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| |
Collapse
|
427
|
|
428
|
Flesken-Nikitin A, Odai-Afotey AA, Nikitin AY. Role of the stem cell niche in the pathogenesis of epithelial ovarian cancers. Mol Cell Oncol 2014; 1:e963435. [PMID: 27308341 PMCID: PMC4905019 DOI: 10.4161/23723548.2014.963435] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 08/02/2014] [Accepted: 08/07/2014] [Indexed: 01/10/2023]
Abstract
Ovarian cancer is the fifth leading cause of cancer-related deaths among women in the United States. Recent extensive genomic analyses of epithelial ovarian cancer (EOC), particularly the most common and deadly form of high-grade serous ovarian carcinoma, have provided important insights into the repertoire of molecular aberrations that are characteristic for this malignancy. However, interpretation of the discovered aberrations is complicated because the origin and mechanisms of progression of EOC remain uncertain. Here, we summarize current views on the cell of origin of EOC and discuss recent findings of a cancer-prone stem cell niche for ovarian surface epithelium, one of the major likely sources of EOC. We also outline future directions and challenges in studying the role of stem cell niches in EOC pathogenesis.
Collapse
Affiliation(s)
- Andrea Flesken-Nikitin
- Department of Biomedical Sciences and Stem Cell Program; Cornell University ; Ithaca, NY USA
| | - Ashley A Odai-Afotey
- Department of Biomedical Sciences and Stem Cell Program; Cornell University ; Ithaca, NY USA
| | - Alexander Yu Nikitin
- Department of Biomedical Sciences and Stem Cell Program; Cornell University ; Ithaca, NY USA
| |
Collapse
|
429
|
Peregrina K, Houston M, Daroqui C, Dhima E, Sellers RS, Augenlicht LH. Vitamin D is a determinant of mouse intestinal Lgr5 stem cell functions. Carcinogenesis 2014; 36:25-31. [PMID: 25344836 DOI: 10.1093/carcin/bgu221] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Lgr5+ intestinal crypt base columnar cells function as stem cells whose progeny populate the villi, and Lgr5+ cells in which Apc is inactivated can give rise to tumors. Surprisingly, these Lgr5+ stem cell properties were abrogated by the lower dietary vitamin D and calcium in a semi-purified diet that promotes both genetically initiated and sporadic intestinal tumors. Inactivation of the vitamin D receptor in Lgr5+ cells established that compromise of Lgr5 stem cell function was a rapid, cell autonomous effect of signaling through the vitamin D receptor. The loss of Lgr5 stem cell function was associated with presence of Ki67 negative Lgr5+ cells at the crypt base. Therefore, vitamin D, a common nutrient and inducer of intestinal cell maturation, is an environmental factor that is a determinant of Lgr5+ stem cell functions in vivo. Since diets used in reports that establish and dissect mouse Lgr5+ stem cell activity likely provided vitamin D levels well above the range documented for human populations, the contribution of Lgr5+ cells to intestinal homeostasis and tumor formation in humans may be significantly more limited, and variable in the population, then suggested by published rodent studies.
Collapse
Affiliation(s)
- Karina Peregrina
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Michele Houston
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Cecilia Daroqui
- Clinica Reina Fabiola, Oncativo 1248, Cordoba 5004, Argentina
| | - Elena Dhima
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Leonard H Augenlicht
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA, Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
430
|
BLIMP1 is required for postnatal epidermal homeostasis but does not define a sebaceous gland progenitor under steady-state conditions. Stem Cell Reports 2014; 3:620-33. [PMID: 25358790 PMCID: PMC4223714 DOI: 10.1016/j.stemcr.2014.08.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 08/08/2014] [Accepted: 08/11/2014] [Indexed: 01/03/2023] Open
Abstract
B-lymphocyte-induced nuclear maturation protein 1 (BLIMP1) was previously reported to define a sebaceous gland (SG) progenitor population in the epidermis. However, the recent identification of multiple stem cell populations in the hair follicle junctional zone has led us to re-evaluate its function. We show, in agreement with previous studies, that BLIMP1 is expressed by postmitotic, terminally differentiated epidermal cells within the SG, interfollicular epidermis, and hair follicle. Epidermal overexpression of c-Myc results in loss of BLIMP1(+) cells, an effect modulated by androgen signaling. Epidermal-specific deletion of Blimp1 causes multiple differentiation defects in the epidermis in addition to SG enlargement. In culture, BLIMP1(+) sebocytes have no greater clonogenic potential than BLIMP1(-) sebocytes. Finally, lineage-tracing experiments reveal that, under steady-state conditions, BLIMP1-expressing cells do not divide. Thus, rather than defining a sebocyte progenitor population, BLIMP1 functions in terminally differentiated cells to maintain homeostasis in multiple epidermal compartments.
Collapse
|
431
|
Hogan BLM, Barkauskas CE, Chapman HA, Epstein JA, Jain R, Hsia CCW, Niklason L, Calle E, Le A, Randell SH, Rock J, Snitow M, Krummel M, Stripp BR, Vu T, White ES, Whitsett JA, Morrisey EE. Repair and regeneration of the respiratory system: complexity, plasticity, and mechanisms of lung stem cell function. Cell Stem Cell 2014; 15:123-38. [PMID: 25105578 PMCID: PMC4212493 DOI: 10.1016/j.stem.2014.07.012] [Citation(s) in RCA: 652] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Respiratory disease is the third leading cause of death in the industrialized world. Consequently, the trachea, lungs, and cardiopulmonary vasculature have been the focus of extensive investigations. Recent studies have provided new information about the mechanisms driving lung development and differentiation. However, there is still much to learn about the ability of the adult respiratory system to undergo repair and to replace cells lost in response to injury and disease. This Review highlights the multiple stem/progenitor populations in different regions of the adult lung, the plasticity of their behavior in injury models, and molecular pathways that support homeostasis and repair.
Collapse
Affiliation(s)
- Brigid L M Hogan
- Department of Cell Biology, Duke Medicine, Durham, NC 27705, USA.
| | - Christina E Barkauskas
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke Medicine, Durham, NC 27705, USA
| | - Harold A Chapman
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jonathan A Epstein
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rajan Jain
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Connie C W Hsia
- Department of Internal Medicine, University of Texas, Southwestern Medical Center, Dallas, TX 75390, USA
| | - Laura Niklason
- Departments of Anesthesiology and Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Elizabeth Calle
- Department of Cell Biology, Duke Medicine, Durham, NC 27705, USA
| | - Andrew Le
- Department of Cell Biology, Duke Medicine, Durham, NC 27705, USA
| | - Scott H Randell
- Department of Cell Biology and Physiology, The University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jason Rock
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Melinda Snitow
- Perleman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew Krummel
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Barry R Stripp
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Thiennu Vu
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eric S White
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeffrey A Whitsett
- Section of Neonatology, Perinatal and Pulmonary Biology, Department of Pediatrics, Cincinnati Children's Hospital Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Edward E Morrisey
- Departments of Medicine and Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|