1
|
Kim YK, Ramalho-Santos M. 20 years of stemness: From stem cells to hypertranscription and back. Stem Cell Reports 2025; 20:102406. [PMID: 39919752 PMCID: PMC11960510 DOI: 10.1016/j.stemcr.2025.102406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 02/09/2025] Open
Abstract
Transcriptional profiling of stem cells came of age at the beginning of the century with the use of microarrays to analyze cell populations in bulk. Since then, stem cell transcriptomics has become increasingly sophisticated, notably with the recent widespread use of single-cell RNA sequencing. Here, we provide a perspective on how an early signature of genes upregulated in embryonic and adult stem cells, identified using microarrays over 20 years ago, serendipitously led to the recent discovery that stem/progenitor cells across organs are in a state of hypertranscription, a global elevation of the transcriptome. Looking back, we find that the 2002 stemness signature is a robust marker of stem cell hypertranscription, even though it was developed well before it was known what hypertranscription meant or how to detect it. We anticipate that studies of stem cell hypertranscription will be rich in novel insights in physiological and disease contexts for years to come.
Collapse
Affiliation(s)
- Yun-Kyo Kim
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto ON M5G 1X5, Canada.
| | - Miguel Ramalho-Santos
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto ON M5T 3L9, Canada; Department of Molecular Genetics, University of Toronto, Toronto ON M5G 1X5, Canada.
| |
Collapse
|
2
|
Regmi B, Brooks SR, Uhlman AJ, Sun HW. RichPathR: a gene set enrichment analysis and visualization tool. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555198. [PMID: 37886446 PMCID: PMC10602030 DOI: 10.1101/2023.08.28.555198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Gene set enrichment analysis (GSEA) is an important step for disease and drug discovery. Genomic, transcriptomics, proteomics and epigenetic analysis of tissue or cells generates gene lists that need to be further investigated in the known biological context. The advent of high-throughput technologies generates the vast number of gene lists that are up or down regulated together. One way of getting meaningful insights of the relationship of these genes is utilizing existing knowledge bases linking them with biological functions or phenotypes. Multiple public databases with annotated gene sets are available for GSEA, and enrichR is the most popular web application still requiring custom tools for large-scale mining. richPathR package is a collection of R functions that helps researchers carry out exploratory analysis and visualization of gene set enrichment using EnrichR.
Collapse
Affiliation(s)
- Binod Regmi
- Biodata Mining and Discovery Section, NIAMS, NIH, Bethesda, MD, USA 1
| | - Stephen R. Brooks
- Biodata Mining and Discovery Section, NIAMS, NIH, Bethesda, MD, USA 1
| | - Andrew J. Uhlman
- Biodata Mining and Discovery Section, NIAMS, NIH, Bethesda, MD, USA 1
| | - Hong-Wei Sun
- Biodata Mining and Discovery Section, NIAMS, NIH, Bethesda, MD, USA 1
| |
Collapse
|
3
|
Hegde M, Girisa S, Kunnumakkara AB. A compilation of bioinformatic approaches to identify novel downstream targets for the detection and prophylaxis of cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 134:75-113. [PMID: 36858743 DOI: 10.1016/bs.apcsb.2022.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
The paradigm of cancer genomics has been radically changed by the development in next-generation sequencing (NGS) technologies making it possible to envisage individualized treatment based on tumor and stromal cells genome in a clinical setting within a short timeframe. The abundance of data has led to new avenues for studying coordinated alterations that impair biological processes, which in turn has increased the demand for bioinformatic tools for pathway analysis. While most of this work has been concentrated on optimizing certain algorithms to obtain quicker and more accurate results. Large volumes of these existing algorithm-based data are difficult for the biologists and clinicians to access, download and reanalyze them. In the present study, we have listed the bioinformatics algorithms and user-friendly graphical user interface (GUI) tools that enable code-independent analysis of big data without compromising the quality and time. We have also described the advantages and drawbacks of each of these platforms. Additionally, we emphasize the importance of creating new, more user-friendly solutions to provide better access to open data and talk about relevant problems like data sharing and patient privacy.
Collapse
Affiliation(s)
- Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, India
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, India.
| |
Collapse
|
4
|
Baranovsky A, Ivanov T, Granovskaya M, Papatsenko D, Pervouchine DD. Transcriptome analysis reveals high tumor heterogeneity with respect to re-activation of stemness and proliferation programs. PLoS One 2022; 17:e0268626. [PMID: 35587924 PMCID: PMC9119523 DOI: 10.1371/journal.pone.0268626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 05/03/2022] [Indexed: 12/01/2022] Open
Abstract
Significant alterations in signaling pathways and transcriptional regulatory programs together represent major hallmarks of many cancers. These, among all, include the reactivation of stemness, which is registered by the expression of pathways that are active in the embryonic stem cells (ESCs). Here, we assembled gene sets that reflect the stemness and proliferation signatures and used them to analyze a large panel of RNA-seq data from The Cancer Genome Atlas (TCGA) Consortium in order to specifically assess the expression of stemness-related and proliferation-related genes across a collection of different tumor types. We introduced a metric that captures the collective similarity of the expression profile of a tumor to that of ESCs, which showed that stemness and proliferation signatures vary greatly between different tumor types. We also observed a high degree of intertumoral heterogeneity in the expression of stemness- and proliferation-related genes, which was associated with increased hazard ratios in a fraction of tumors and mirrored by high intratumoral heterogeneity and a remarkable stemness capacity in metastatic lesions across cancer cells in single cell RNA-seq datasets. Taken together, these results indicate that the expression of stemness signatures is highly heterogeneous and cannot be used as a universal determinant of cancer. This calls into question the universal validity of diagnostic tests that are based on stem cell markers.
Collapse
Affiliation(s)
- Artem Baranovsky
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
- Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Timofei Ivanov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
| | | | - Dmitri Papatsenko
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Dmitri D. Pervouchine
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
- * E-mail:
| |
Collapse
|
5
|
Modvig S, Wernersson R, Øbro NF, Olsen LR, Christensen C, Rosthøj S, Degn M, Jürgensen GW, Madsen HO, Albertsen BK, Wehner PS, Rosthøj S, Lilljebjörn H, Fioretos T, Schmiegelow K, Marquart HV. High CD34 surface expression in BCP-ALL predicts poor induction therapy response and is associated with altered expression of genes related to cell migration and adhesion. Mol Oncol 2022; 16:2015-2030. [PMID: 35271751 PMCID: PMC9120905 DOI: 10.1002/1878-0261.13207] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/01/2022] [Accepted: 03/07/2022] [Indexed: 11/25/2022] Open
Abstract
Minimal residual disease (MRD) constitutes the most important prognostic factor in B‐cell precursor acute lymphoblastic leukemia (BCP‐ALL). Flow cytometry is widely used in MRD assessment, yet little is known regarding the effect of different immunophenotypic subsets on outcome. In this study of 200 BCP‐ALL patients, we found that a CD34‐positive, CD38 dim‐positive, nTdT dim‐positive immunophenotype on the leukemic blasts was associated with poor induction therapy response and predicted an MRD level at the end of induction therapy (EOI) of ≥ 0.001. CD34 expression was strongly and positively associated with EOI MRD, whereas CD34‐negative patients had a low relapse risk. Further, CD34 expression increased from diagnosis to relapse. CD34 is a stemness‐associated cell‐surface molecule, possibly involved in cell adhesion/migration or survival. Accordingly, genes associated with stemness were overrepresented among the most upregulated genes in CD34‐positive leukemias, and protein–protein interaction networks showed an overrepresentation of genes associated with cell migration, cell adhesion, and negative regulation of apoptosis. The present work is the first to demonstrate a CD34‐negative immunophenotype as a good prognostic factor in ALL, whereas high CD34 expression is associated with poor therapy response and an altered gene expression profile reminiscent of migrating cancer stem‐like cells.
Collapse
Affiliation(s)
- Signe Modvig
- Dept. of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Rasmus Wernersson
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark.,Intomics A/S, Lyngby, Denmark
| | - Nina Friesgaard Øbro
- Dept. of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Lars Rønn Olsen
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Claus Christensen
- Dept. of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Susanne Rosthøj
- Section of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Matilda Degn
- Dept. of Pediatric and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet
| | - Gitte Wullf Jürgensen
- Dept. of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Hans O Madsen
- Dept. of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Birgitte Klug Albertsen
- Dept. of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Peder Skov Wehner
- H.C. Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| | - Steen Rosthøj
- Department of Pediatrics and Adolescent Medicine, Aalborg University Hospital, Aalborg, Denmark
| | - Henrik Lilljebjörn
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Thoas Fioretos
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Kjeld Schmiegelow
- Dept. of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Dept. of Pediatric and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Faculty of Medicine, Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Hanne Vibeke Marquart
- Dept. of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
6
|
Du J, Liu X, Yarema KJ, Jia X. Glycoengineering human neural stem cells (hNSCs) for adhesion improvement using a novel thiol-modified N-acetylmannosamine (ManNAc) analog. BIOMATERIALS ADVANCES 2022; 134:112675. [PMID: 35599100 PMCID: PMC9300770 DOI: 10.1016/j.msec.2022.112675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/20/2022]
Abstract
This study sets the stage for the therapeutic use of Ac5ManNTProp, an N-acetylmannosamine (ManNAc) analog that installs thiol-modified sialoglycans onto the surfaces of human neural stem cells (hNSC). First, we compared hNSC adhesion to the extracellular matrix (ECM) proteins laminin, fibronectin, and collagen and found preferential adhesion and concomitant changes to cell morphology and cell spreading for Ac5ManNTProp-treated cells to laminin, compared to fibronectin where there was a modest response, and collagen where there was no observable increase. PCR array transcript analysis identified several classes of cell adhesion molecules that responded to combined Ac5ManNTProp treatment and hNSC adhesion to laminin. Of these, we focused on integrin α6β1 expression, which was most strongly upregulated in analog-treated cells incubated on laminin. We also characterized downstream responses including vinculin display as well as the phosphorylation of focal adhesion kinase (FAK) and extracellular signal-related kinase (ERK). In these experiments, Ac5ManNTProp more strongly induced all tested biological endpoints compared to Ac5ManNTGc, showing that the single methylene unit that structurally separates the two analogs finely tunes biological responses. Together, the concerted modulation of multiple pro-regenerative activities through Ac5ManNTProp treatment, in concert with crosstalk with ECM components, lays a foundation for using our metabolic glycoengineering approach to treat neurological disorders by favorably modulating endpoints that contribute to the viability of transplanted NSCs.
Collapse
Affiliation(s)
- Jian Du
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Xiao Liu
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Kevin J. Yarema
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, 21205,Translational Cell and Tissue Engineering Center, The Johns Hopkins School of Medicine, Baltimore, MD, 21231
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
7
|
Weerakoon H, Straube J, Lineburg K, Cooper L, Lane S, Smith C, Alabbas S, Begun J, Miles JJ, Hill MM, Lepletier A. Expression of CD49f defines subsets of human regulatory T cells with divergent transcriptional landscape and function that correlate with ulcerative colitis disease activity. Clin Transl Immunology 2021; 10:e1334. [PMID: 34504692 PMCID: PMC8419695 DOI: 10.1002/cti2.1334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/04/2021] [Accepted: 08/05/2021] [Indexed: 01/08/2023] Open
Abstract
Objective Adoptive regulatory T cell (Treg) therapy is being trialled for the treatment of different autoimmune disorders, including inflammatory bowel diseases (IBD). In‐depth understanding of the biological variability of Treg in the human blood may be required to improve IBD immune monitoring and treatment strategies. Methods Through a combination of quantitative proteomic, multiparametric flow cytometry, RNA‐sequencing data analysis and functional assays on Treg enriched from the blood of ulcerative colitis (UC) patients and healthy controls, we investigated the association between CD49f expression, Treg phenotype and function, and UC disease activity. Results High‐dimensional analysis and filtering defined two distinct subsets of human Treg based on the presence or absence of CD49f with divergent transcriptional landscape and functional activities. CD49f negative (CD49f−) Treg are enriched for functional Treg markers and present significantly increased suppressive capacity. In contrast, CD49fhigh Treg display a pro‐inflammatory Th17‐like phenotype and accumulate in the blood of patients with UC. Dysregulation on CD49f Treg subsets in patients with UC correlate with disease activity. Conclusion Overall, our findings uncover the importance of CD49f expression on Treg in physiological immunity and in pathological autoimmunity.
Collapse
Affiliation(s)
- Harshi Weerakoon
- Precision and Systems Biomedicine Laboratory QIMR Berghofer Medical Research Institute Herston QLD Australia.,School of Biomedical Sciences The University of Queensland Brisbane QLD Australia.,Department of Biochemistry Faculty of Medicine and Allied Sciences Rajarata University of Sri Lanka Saliyapura Sri Lanka
| | - Jasmin Straube
- Gordon and Jessie Gilmour Leukaemia Research Laboratory QIMR Berghofer Medical Research Institute Herston QLD Australia
| | - Katie Lineburg
- Translational and Human Immunology Laboratory QIMR Berghofer Medical Research Institute Herston QLD Australia
| | - Leanne Cooper
- Gordon and Jessie Gilmour Leukaemia Research Laboratory QIMR Berghofer Medical Research Institute Herston QLD Australia
| | - Steven Lane
- Gordon and Jessie Gilmour Leukaemia Research Laboratory QIMR Berghofer Medical Research Institute Herston QLD Australia.,School of Medicine University of Queensland Brisbane QLD Australia
| | - Corey Smith
- Translational and Human Immunology Laboratory QIMR Berghofer Medical Research Institute Herston QLD Australia
| | - Saleh Alabbas
- Inflammatory Bowel Diseases Research Group Mater Research Institute University of Queensland Brisbane QLD Australia
| | - Jakob Begun
- School of Medicine University of Queensland Brisbane QLD Australia.,Inflammatory Bowel Diseases Research Group Mater Research Institute University of Queensland Brisbane QLD Australia.,Mater Hospital Brisbane Brisbane QLD Australia
| | - John J Miles
- Human Immunity Laboratory QIMR Berghofer Medical Research Institute Herston QLD Australia.,Centre for Biodiscovery and Molecular Development of Therapeutics James Cook University Cairns QLD Australia
| | - Michelle M Hill
- Precision and Systems Biomedicine Laboratory QIMR Berghofer Medical Research Institute Herston QLD Australia.,Centre for Clinical Research Faculty of Medicine The University of Queensland Brisbane QLD Australia
| | - Ailin Lepletier
- Human Immunity Laboratory QIMR Berghofer Medical Research Institute Herston QLD Australia.,Laboratory of Vaccines for the Developing World Institute for Glycomics Southport QLD Australia
| |
Collapse
|
8
|
Parker KA, Gooding AJ, Valadkhan S, Schiemann WP. lncRNA BORG:TRIM28 Complexes Drive Metastatic Progression by Inducing α6 Integrin/CD49f Expression in Breast Cancer Stem Cells. Mol Cancer Res 2021; 19:2068-2080. [PMID: 34497119 DOI: 10.1158/1541-7786.mcr-21-0137] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/22/2021] [Accepted: 08/27/2021] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most lethal subtype of breast cancer, with its aggressive phenotype being attributed to chemotherapy resistance, metastatic dissemination, and rapid disease recurrence. Breast cancer stem cells (BCSC) are significant contributors to tumor initiation, as well as to the acquisition of aggressive tumorigenic phenotypes, namely due to their ability to self-replicate and to produce heterogeneous differentiated tumor cells. To elucidate the underlying mechanisms that drive BCSC tumorigenicity in TNBC, we identified the long noncoding RNA (lncRNA) B MP/ O P- R esponsive G ene (BORG) as an enhancer of BCSC phenotypes. Indeed, we found BORG expression to: (i) correlate with stem cell markers Nanog, Aldh1a3, and Itga6 (α6 integrin/CD49f); (ii) enhance stem cell phenotypes in murine and human TNBC cells, and (iii) promote TNBC tumor initiation in mice. Mechanistically, BORG promoted BCSC phenotypes through its ability to interact physically with the E3 SUMO ligase TRIM28. Moreover, TRIM28 binding was observed in the promoter region of Itga6, whose genetic inactivation prevented BORG:TRIM28 complexes from: (i) inducing BCSC self-renewal and expansion in vitro, and (ii) eliciting BCSC metastatic outgrowth in the lungs of mice. Collectively, these findings implicate BORG:TRIM28 complexes as novel drivers of BCSC phenotypes in developing and progressing TNBCs. IMPLICATIONS: This work establishes the lncRNA BORG as a driver of BCSC phenotypes and the aggressive behaviors of TNBCs, events critically dependent upon the formation of BORG:TRIM28 complexes and expression of α6 integrin.
Collapse
Affiliation(s)
- Kimberly A Parker
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio
| | - Alex J Gooding
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Saba Valadkhan
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio
| | - William P Schiemann
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
9
|
Ibrahim MR, Medhat W, El-Fakahany H, Abdel-Raouf H, Snyder EY. The Developmental & Molecular Requirements for Ensuring that Human Pluripotent Stem Cell-Derived Hair Follicle Bulge Stem Cells Have Acquired Competence for Hair Follicle Generation Following Transplantation. Cell Transplant 2021; 30:9636897211014820. [PMID: 34053245 PMCID: PMC8182633 DOI: 10.1177/09636897211014820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
When using human induced pluripotent stem cells (hiPSCs) to achieve hair follicle (HF) replacement, we found it best to emulate the earliest fundamental developmental processes of gastrulation, ectodermal lineage commitment, and dermogenesis. Viewing hiPSCs as a model of the epiblast, we exploited insights from mapping the dynamic up- and down-regulation of the developmental molecules that determine HF lineage in order to ascertain the precise differentiation stage and molecular requirements for grafting HF-generating progenitors. To yield an integrin-dependent lineage like the HF in vivo, we show that hiPSC derivatives should co-express, just prior to transplantation, the following combination of markers: integrins α6 and β1 and the glycoprotein CD200 on their surface; and, intracellularly, the epithelial marker keratin 18 and the hair follicle bulge stem cell (HFBSC)-defining molecules transcription factor P63 and the keratins 15 and 19. If the degree of trichogenic responsiveness indicated by the presence of these molecules is not achieved (they peak on Days 11-18 of the protocol), HF generation is not possible. Conversely, if differentiation of the cells is allowed to proceed beyond the transient intermediate progenitor state represented by the HFBSC, and instead cascades to their becoming keratin 14+ keratin 5+ CD200– keratinocytes (Day 25), HF generation is equally impossible. We make the developmental case for transplanting at Day 16-18 of differentiation—the point at which the hiPSCs have lost pluripotency, have attained optimal expression of HFBSC markers, have not yet experienced downregulation of key integrins and surface glycoproteins, have not yet started expressing keratinocyte-associated molecules, and have sufficient proliferative capacity to allow a well-populated graft. This panel of markers may be used for isolating (by cytometry) HF-generating derivatives away from cell types unsuited for this therapy as well as for identifying trichogenic drugs.
Collapse
Affiliation(s)
- Michel R Ibrahim
- Department of Dermatology, STD's and Andrology, Faculty of Medicine, Minia University, Al-Minya, Egypt.,Center for Stem Cells & Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Walid Medhat
- Department of Dermatology, STD's and Andrology, Faculty of Medicine, Minia University, Al-Minya, Egypt
| | - Hasan El-Fakahany
- Department of Dermatology, STD's and Andrology, Faculty of Medicine, Minia University, Al-Minya, Egypt
| | - Hamza Abdel-Raouf
- Department of Dermatology, STD's and Andrology, Faculty of Medicine, Minia University, Al-Minya, Egypt
| | - Evan Y Snyder
- Center for Stem Cells & Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA.,Department of Pediatrics, University of California-San Diego, La Jolla, CA, USA
| |
Collapse
|
10
|
When the Search for Stemness Genes Meets the Skin Substitute Bioengineering Field: KLF4 Transcription Factor under the Light. Cells 2020; 9:cells9102188. [PMID: 32998444 PMCID: PMC7601001 DOI: 10.3390/cells9102188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 11/16/2022] Open
Abstract
The transcription factor “Kruppel-like factor 4” (KLF4) is a central player in the field of pluripotent stem cell biology. In particular, it was put under the spotlight as one of the four factors of the cocktail originally described for reprogramming into induced pluripotent stem cells (iPSCs). In contrast, its possible functions in native tissue stem cells remain largely unexplored. We recently published that KLF4 is a regulator of “stemness” in human keratinocytes. We show that reducing the level of expression of this transcription factor by RNA interference or pharmacological repression promotes the ex vivo amplification and regenerative capacity of two types of cells of interest for cutaneous cell therapy: native keratinocyte stem and progenitor cells from adult epidermis, which have been used for more than three decades in skin graft bioengineering, and keratinocytes generated by the lineage-oriented differentiation of embryonic stem cells (ESCs), which have potential for the development of skin bio-bandages. At the mechanistic level, KLF4 repression alters the expression of a large set of genes involved in TGF-β1 and WNT signaling pathways. Major regulators of TGF-β bioavailability and different TGF-β receptors were targeted, notably modulating the ALK1/Smad1/5/9 axis. At a functional level, KLF4 repression produced an antagonist effect on TGF-β1-induced keratinocyte differentiation.
Collapse
|
11
|
Pradines JR, Farutin V, Cilfone NA, Ghavami A, Kurtagic E, Guess J, Manning AM, Capila I. Enhancing reproducibility of gene expression analysis with known protein functional relationships: The concept of well-associated protein. PLoS Comput Biol 2020; 16:e1007684. [PMID: 32058996 PMCID: PMC7046299 DOI: 10.1371/journal.pcbi.1007684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 02/27/2020] [Accepted: 01/27/2020] [Indexed: 12/27/2022] Open
Abstract
Identification of differentially expressed genes (DEGs) is well recognized to be variable across independent replications of genome-wide transcriptional studies. These are often employed to characterize disease state early in the process of discovery and prioritize novel targets aimed at addressing unmet medical need. Increasing reproducibility of biological findings from these studies could potentially positively impact the success rate of new clinical interventions. This work demonstrates that statistically sound combination of gene expression data with prior knowledge about biology in the form of large protein interaction networks can yield quantitatively more reproducible observations from studies characterizing human disease. The novel concept of Well-Associated Proteins (WAPs) introduced herein-gene products significantly associated on protein interaction networks with the differences in transcript levels between control and disease-does not require choosing a differential expression threshold and can be computed efficiently enough to enable false discovery rate estimation via permutation. Reproducibility of WAPs is shown to be on average superior to that of DEGs under easily-quantifiable conditions suggesting that they can yield a significantly more robust description of disease. Enhanced reproducibility of WAPs versus DEGs is first demonstrated with four independent data sets focused on systemic sclerosis. This finding is then validated over thousands of pairs of data sets obtained by random partitions of large studies in several other diseases. Conditions that individual data sets must satisfy to yield robust WAP scores are examined. Reproducible identification of WAPs can potentially benefit drug target selection and precision medicine studies.
Collapse
Affiliation(s)
- Joël R. Pradines
- Momenta Pharmaceuticals, 301 Binney Street, Cambridge, Massachusetts, United States of America
| | - Victor Farutin
- Momenta Pharmaceuticals, 301 Binney Street, Cambridge, Massachusetts, United States of America
- * E-mail: (VF); (IC)
| | - Nicholas A. Cilfone
- Momenta Pharmaceuticals, 301 Binney Street, Cambridge, Massachusetts, United States of America
| | - Abouzar Ghavami
- Momenta Pharmaceuticals, 301 Binney Street, Cambridge, Massachusetts, United States of America
| | - Elma Kurtagic
- Momenta Pharmaceuticals, 301 Binney Street, Cambridge, Massachusetts, United States of America
| | - Jamey Guess
- Momenta Pharmaceuticals, 301 Binney Street, Cambridge, Massachusetts, United States of America
| | - Anthony M. Manning
- Momenta Pharmaceuticals, 301 Binney Street, Cambridge, Massachusetts, United States of America
| | - Ishan Capila
- Momenta Pharmaceuticals, 301 Binney Street, Cambridge, Massachusetts, United States of America
- * E-mail: (VF); (IC)
| |
Collapse
|
12
|
Li QV, Rosen BP, Huangfu D. Decoding pluripotency: Genetic screens to interrogate the acquisition, maintenance, and exit of pluripotency. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1464. [PMID: 31407519 PMCID: PMC6898739 DOI: 10.1002/wsbm.1464] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 05/31/2019] [Accepted: 07/17/2019] [Indexed: 01/25/2023]
Abstract
Pluripotent stem cells have the ability to unlimitedly self-renew and differentiate to any somatic cell lineage. A number of systems biology approaches have been used to define this pluripotent state. Complementary to systems level characterization, genetic screens offer a unique avenue to functionally interrogate the pluripotent state and identify the key players in pluripotency acquisition and maintenance, exit of pluripotency, and lineage differentiation. Here we review how genetic screens have helped us decode pluripotency regulation. We will summarize results from RNA interference (RNAi) based screens, discuss recent advances in CRISPR/Cas-based genetic perturbation methods, and how these advances have made it possible to more comprehensively interrogate pluripotency and differentiation through genetic screens. Such investigations will not only provide a better understanding of this unique developmental state, but may enhance our ability to use pluripotent stem cells as an experimental model to study human development and disease progression. Functional interrogation of pluripotency also provides a valuable roadmap for utilizing genetic perturbation to gain systems level understanding of additional cellular states, from later stages of development to pathological disease states. This article is categorized under: Developmental Biology > Stem Cell Biology and Regeneration Developmental Biology > Developmental Processes in Health and Disease Biological Mechanisms > Cell Fates.
Collapse
Affiliation(s)
- Qing V. Li
- Sloan Kettering Institute, 1275 York Avenue, New York, New York 10065, USA
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
- These authors contributed equally
| | - Bess P. Rosen
- Sloan Kettering Institute, 1275 York Avenue, New York, New York 10065, USA
- Weill Graduate School of Medical Sciences at Cornell University, 1300 York Avenue, New York, New York 10065, USA
- These authors contributed equally
| | - Danwei Huangfu
- Sloan Kettering Institute, 1275 York Avenue, New York, New York 10065, USA
| |
Collapse
|
13
|
Fortunel NO, Chadli L, Coutier J, Lemaître G, Auvré F, Domingues S, Bouissou-Cadio E, Vaigot P, Cavallero S, Deleuze JF, Roméo PH, Martin MT. KLF4 inhibition promotes the expansion of keratinocyte precursors from adult human skin and of embryonic-stem-cell-derived keratinocytes. Nat Biomed Eng 2019; 3:985-997. [PMID: 31636412 DOI: 10.1038/s41551-019-0464-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 09/13/2019] [Indexed: 01/01/2023]
Abstract
Expanded autologous skin keratinocytes are currently used in cutaneous cell therapy, and embryonic-stem-cell-derived keratinocytes could become a complementary alternative. Regardless of keratinocyte provenance, for efficient therapy it is necessary to preserve immature keratinocyte precursors during cell expansion and graft processing. Here, we show that stable and transient downregulation of the transcription factor Krüppel-like factor 4 (KLF4) in keratinocyte precursors from adult skin, using anti-KLF4 RNA interference or kenpaullone, promotes keratinocyte immaturity and keratinocyte self-renewal in vitro, and enhances the capacity for epidermal regeneration in mice. Both stable and transient KLF4 downregulation had no impact on the genomic integrity of adult keratinocytes. Moreover, transient KLF4 downregulation in human-embryonic-stem-cell-derived keratinocytes increased the efficiency of skin-orientated differentiation and of keratinocyte immaturity, and was associated with improved generation of epidermis. As a regulator of the cell fate of keratinocyte precursors, KLF4 could be used for promoting the ex vivo expansion and maintenance of functional immature keratinocyte precursors.
Collapse
Affiliation(s)
- Nicolas O Fortunel
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France. .,INSERM U967, Université Paris-Diderot, Paris, France. .,Université Paris-Saclay, Paris, France.
| | - Loubna Chadli
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Université Paris-Diderot, Paris, France.,Université Paris-Saclay, Paris, France
| | - Julien Coutier
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Université Paris-Diderot, Paris, France.,Université Paris-Saclay, Paris, France
| | - Gilles Lemaître
- Université d'Evry Val d'Essonne, Université Paris-Saclay, INSERM U861, Institut des Cellules Souches pour le Traitement et l'Etude des Maladies Monogéniques, Corbeil Essonne, France
| | - Frédéric Auvré
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Université Paris-Diderot, Paris, France.,Université Paris-Saclay, Paris, France
| | - Sophie Domingues
- Centre d'Etude des Cellules Souches, Institut des Cellules Souches pour le Traitement et l'Etude des Maladies Monogéniques, Corbeil Essonne, France
| | - Emmanuelle Bouissou-Cadio
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Université Paris-Diderot, Paris, France.,Université Paris-Saclay, Paris, France
| | - Pierre Vaigot
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Université Paris-Diderot, Paris, France.,Université Paris-Saclay, Paris, France
| | - Sophie Cavallero
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Université Paris-Diderot, Paris, France.,Université Paris-Saclay, Paris, France
| | | | - Paul-Henri Roméo
- INSERM U967, Université Paris-Diderot, Paris, France.,Université Paris-Saclay, Paris, France.,Laboratoire de Recherche sur la Réparation et la Transcription dans les Cellules Souches, CEA/DRF/IBFJ/IRCM, Fontenay-aux-Roses, France
| | - Michèle T Martin
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France. .,INSERM U967, Université Paris-Diderot, Paris, France. .,Université Paris-Saclay, Paris, France.
| |
Collapse
|
14
|
Zhou L, Che Z, Zhang X, Zhou P, Li X, Xu X, Shi Q, Li D, Ju X. Influence of neonatal gender on cord blood CD34 + cell amplification and gene expression. Exp Ther Med 2019; 18:105-118. [PMID: 31258643 PMCID: PMC6566080 DOI: 10.3892/etm.2019.7549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 03/12/2019] [Indexed: 12/18/2022] Open
Abstract
The present study attempted to evaluate whether neonatal gender affects the hematopoietic potential of cord blood (CB) transplants and, if so, to determine the underlying molecular mechanisms. CD34+ cells from CB were isolated and divided into male and female groups. CD34+CD38− cell populations were then compared using fluorescence-assisted cell sorting (FACS) and a colony formation assay was performed. Next, a Genechip microarray analysis was used to identify differentially expressed genes (DEGs). Finally, the Genechip results were validated by FACS analysis. It was revealed that the male group had higher amplification efficiency. Gene ontology analysis indicated differences in the biological function of the DEGs between the two groups. Kyoto Encyclopedia of Genes and Genomes analysis suggested that the hematopoietic cell lineage signaling pathway was upregulated in the male group along with high expression levels of genes including interleukin (IL) 6 signal transducer (glycoprotein 130), IL-7 and IL-7 receptor. It was speculated that this may be partially due to numerous upregulated DEGs being involved in chromosomal segregation and hematopoietic cell lineage signaling pathways in CD34+ cells from the male group.
Collapse
Affiliation(s)
- Liping Zhou
- Department of Pediatrics, The Sixth People's Hospital of Ji'nan, Jinan, Shandong 250200, P.R. China.,Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhe Che
- Health and Family Planning Inspection Agency of Zhangqiu District, Jinan, Shandong 250200, P.R. China
| | - Xiaowei Zhang
- Department of Pediatrics, The Sixth People's Hospital of Ji'nan, Jinan, Shandong 250200, P.R. China
| | - Panpan Zhou
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xue Li
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xuejing Xu
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qing Shi
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Dong Li
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,Stem Cell and Regenerative Medicine Research Center, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiuli Ju
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,Stem Cell and Regenerative Medicine Research Center, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
15
|
Vassalli G. Aldehyde Dehydrogenases: Not Just Markers, but Functional Regulators of Stem Cells. Stem Cells Int 2019; 2019:3904645. [PMID: 30733805 PMCID: PMC6348814 DOI: 10.1155/2019/3904645] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/25/2018] [Indexed: 12/26/2022] Open
Abstract
Aldehyde dehydrogenase (ALDH) is a superfamily of enzymes that detoxify a variety of endogenous and exogenous aldehydes and are required for the biosynthesis of retinoic acid (RA) and other molecular regulators of cellular function. Over the past decade, high ALDH activity has been increasingly used as a selectable marker for normal cell populations enriched in stem and progenitor cells, as well as for cell populations from cancer tissues enriched in tumor-initiating stem-like cells. Mounting evidence suggests that ALDH not only may be used as a marker for stem cells but also may well regulate cellular functions related to self-renewal, expansion, differentiation, and resistance to drugs and radiation. ALDH exerts its functional actions partly through RA biosynthesis, as all-trans RA reverses the functional effects of pharmacological inhibition or genetic suppression of ALDH activity in many cell types in vitro. There is substantial evidence to suggest that the role of ALDH as a stem cell marker comes down to the specific isoform(s) expressed in a particular tissue. Much emphasis has been placed on the ALDH1A1 and ALDH1A3 members of the ALDH1 family of cytosolic enzymes required for RA biosynthesis. ALDH1A1 and ALDH1A3 regulate cellular function in both normal stem cells and tumor-initiating stem-like cells, promoting tumor growth and resistance to drugs and radiation. An improved understanding of the molecular mechanisms by which ALDH regulates cellular function will likely open new avenues in many fields, especially in tissue regeneration and oncology.
Collapse
Affiliation(s)
- Giuseppe Vassalli
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Lugano, Switzerland
- Center for Molecular Cardiology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
16
|
Zhang Y, Zoltan M, Riquelme E, Xu H, Sahin I, Castro-Pando S, Montiel MF, Chang K, Jiang Z, Ling J, Gupta S, Horne W, Pruski M, Wang H, Sun SC, Lozano G, Chiao P, Maitra A, Leach SD, Kolls JK, Sanchez EV, Wang TC, Bailey JM, McAllister F. Immune Cell Production of Interleukin 17 Induces Stem Cell Features of Pancreatic Intraepithelial Neoplasia Cells. Gastroenterology 2018; 155:210-223.e3. [PMID: 29604293 PMCID: PMC6035075 DOI: 10.1053/j.gastro.2018.03.041] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 03/10/2018] [Accepted: 03/23/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS Little is known about how the immune system affects stem cell features of pancreatic cancer cells. Immune cells that produce interleukin 17A (IL17A) in the chronically inflamed pancreas (chronic pancreatitis) contribute to pancreatic interepithelial neoplasia (PanIN) initiation and progression. We investigated the effects that IL17A signaling exerts on pancreatic cancer progenitor cells and the clinical relevance of this phenomena. METHODS We performed studies with Mist1Cre;LSLKras;Rosa26mTmG (KCiMist;G) and Kras(G12D);Trp53(R172H);Pdx1-Cre (KPC) mice (which upon tamoxifen induction spontaneously develop PanINs) and control littermates. Some mice were injected with neutralizing antibodies against IL17A or control antibody. Pancreata were collected, PanIN epithelial cells were isolated by flow cytometry based on lineage tracing, and gene expression profiles were compared. We collected cells from pancreatic tumors of KPC mice, incubated them with IL17 or control media, measured expression of genes regulated by IL17 signaling, injected the cancer cells into immune competent mice, and measured tumor growth. IL17A was overexpressed in pancreata of KCiMist mice from an adenoviral vector. Pancreata were collected from all mice and analyzed by histology and immunohistochemistry. Levels of DCLK1 and other proteins were knocked down in KPC pancreatic cancer cells using small interfering or short hairpin RNAs; cells were analyzed by immunoblotting. We obtained 65 pancreatic tumor specimens from patients, analyzed protein levels by immunohistochemistry, and compared results with patient survival times. We also analyzed gene expression levels and patient outcome using The Cancer Genome Atlas database. RESULTS PanIN cells from KCiMist;G mice had a gene expression pattern associated with embryonic stem cells. Mice given injections of IL17-neutralizing antibodies, or with immune cells that did not secrete IL17, lost this expression pattern and had significantly decreased expression of DCLK1 and POU2F3, which regulate tuft cell development. KCiMist mice that overexpressed IL17 formed more PanINs, with more DCLK1-positive cells, than control mice. Pancreatic tumor cells from KPC mice and human Capan-2 cells exposed to IL17A had increased activation of NF-κB and mitogen-activated protein kinase signaling and increased expression of DCLK1 and ALDH1A1 (a marker of embryonic stem cells) compared with cells in control media. These cells also formed tumors faster that cells not exposed to IL17 when they were injected into immunocompetent mice. KPC cells with knockdown of DCLK1 expressed lower levels of ALDH1A1 after incubation with IL17 than cells without knockdown. Expression of the IL17 receptor C was higher in DCLK1-positive PanIN cells from mice compared with DCLK1-negative PanIN cells. In human pancreatic tumor tissues, high levels of DCLK1 associated with a shorter median survival time of patients (17.7 months, compared with 26.6 months of patients whose tumors had low levels of DCLK1). Tumor levels of POU2F3 and LAMC2 were also associated with patient survival time. CONCLUSIONS In studies of mouse and human pancreatic tumors and precursors, we found that immune cell-derived IL17 regulated development of tuft cells and stem cell features of pancreatic cancer cells via increased expression of DCLK1, POU2F3, ALDH1A1, and IL17RC. Strategies to disrupt this pathway might be developed to prevent pancreatic tumor growth and progression.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center. Houston, TX, USA
| | - Michelle Zoltan
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center. Houston, TX, USA
| | - Erick Riquelme
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center. Houston, TX, USA
| | - Hanwen Xu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center. Houston, TX, USA
| | - Ismet Sahin
- Department of Engineering, Texas Southern University, Houston, TX
| | - Susana Castro-Pando
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center. Houston, TX, USA
| | - Maria Fernanda Montiel
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center. Houston, TX, USA
| | - Kyle Chang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center. Houston, TX, USA
| | - Zhengyu Jiang
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| | - Jianhua Ling
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sonal Gupta
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - William Horne
- Richard King Mellon Foundation Institute for Pediatric Research, Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Melissa Pruski
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, University of Texas Health Science Center, Houston, TX
| | - Huamin Wang
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Shao-Cong Sun
- Department of Immunology, University of Texas Health Sciences Center, Houston, TX
| | - Guillermina Lozano
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Paul Chiao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anirban Maitra
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Steven D. Leach
- Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jay K. Kolls
- Richard King Mellon Foundation Institute for Pediatric Research, Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Eduardo Vilar Sanchez
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center. Houston, TX, USA
| | - Timothy C. Wang
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| | - Jennifer M. Bailey
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, University of Texas Health Science Center, Houston, TX
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
17
|
Sattiraju A, Sai KKS, Mintz A. Glioblastoma Stem Cells and Their Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:119-140. [PMID: 29204831 DOI: 10.1007/978-3-319-69194-7_7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glioblastoma (GBM) is the most common primary malignant astrocytoma associated with a poor patient survival. Apart from arising de novo, GBMs also occur due to progression of slower growing grade III astrocytomas. GBM is characterized by extensive hypoxia, angiogenesis, proliferation and invasion. Standard treatment options such as surgical resection, radiation therapy and chemotherapy have increased median patient survival to 14.6 months in adults but recurrent disease arising from treatment resistant cancer cells often results in patient mortality. These treatment resistant cancer cells have been found to exhibit stem cell like properties. Strategies to identify or target these Glioblastoma Stem Cells (GSC) have proven to be unsuccessful so far. Studies on cancer stem cells (CSC) within GBM and other cancers have highlighted the importance of paracrine signaling networks within their microenvironment on the growth and maintenance of CSCs. The study of GSCs and their communication with various cell populations within their microenvironment is therefore not only important to understand the biology of GBMs but also to predict response to therapies and to identify novel targets which could stymy support to treatment resistant cancer cells and prevent disease recurrence. The purpose of this chapter is to introduce the concept of GSCs and to detail the latest findings indicating the role of various cellular subtypes within their microenvironment on their survival, proliferation and differentiation.
Collapse
Affiliation(s)
- Anirudh Sattiraju
- Department of Radiology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | | | - Akiva Mintz
- Department of Radiology, Columbia University College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
18
|
Zhou Z, Qu J, He L, Peng H, Chen P, Zhou Y. α6-Integrin alternative splicing: distinct cytoplasmic variants in stem cell fate specification and niche interaction. Stem Cell Res Ther 2018; 9:122. [PMID: 29720266 PMCID: PMC5930856 DOI: 10.1186/s13287-018-0868-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
α6-Integrin subunit (also known as CD49f) is a stemness signature that has been found on the plasma membrane of more than 30 stem cell populations. A growing body of studies have focused on the critical role of α6-containing integrins (α6β1 and α6β4) in the regulation of stem cell properties, lineage-specific differentiation, and niche interaction. α6-Integrin subunit can be alternatively spliced at the post-transcriptional level, giving rise to divergent isoforms which differ in the cytoplasmic and/or extracellular domains. The cytoplasmic domain of integrins is an important functional part of integrin-mediated signals. Structural changes in the cytoplasmic domain of α6 provide an efficient means for the regulation of stem cell responses to biochemical stimuli and/or biophysical cues in the stem cell niche, thus impacting stem cell fate determination. In this review, we summarize the current knowledge on the structural variants of the α6-integrin subunit and spatiotemporal expression of α6 cytoplasmic variants in embryonic and adult stem/progenitor cells. We highlight the roles of α6 cytoplasmic variants in stem cell fate decision and niche interaction, and discuss the potential mechanisms involved. Understanding of the distinct functions of α6 splicing variants in stem cell biology may inform the rational design of novel stem cell-based therapies for a range of human diseases.
Collapse
Affiliation(s)
- Zijing Zhou
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Tinsley Harrison Tower 437B, 1900 University Blvd, Birmingham, AL 35294 USA
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, 410011 Hunan China
| | - Jing Qu
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Tinsley Harrison Tower 437B, 1900 University Blvd, Birmingham, AL 35294 USA
| | - Li He
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Tinsley Harrison Tower 437B, 1900 University Blvd, Birmingham, AL 35294 USA
| | - Hong Peng
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, 410011 Hunan China
| | - Ping Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, 410011 Hunan China
| | - Yong Zhou
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Tinsley Harrison Tower 437B, 1900 University Blvd, Birmingham, AL 35294 USA
| |
Collapse
|
19
|
Auvré F, Coutier J, Martin MT, Fortunel NO. Quantitative Detection of Low-Abundance Transcripts at Single-Cell Level in Human Epidermal Keratinocytes by Digital Droplet Reverse Transcription-Polymerase Chain Reaction. Methods Mol Biol 2018; 1879:31-41. [PMID: 29736807 DOI: 10.1007/7651_2018_149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Genetic and epigenetic characterization of the large cellular diversity observed within tissues is essential to understanding the molecular networks that ensure the regulation of homeostasis, repair, and regeneration, but also pathophysiological processes. Skin is composed of multiple cell lineages and is therefore fully concerned by this complexity. Even within one particular lineage, such as epidermal keratinocytes, different immaturity statuses or differentiation stages are represented, which are still incompletely characterized. Accordingly, there is presently great demand for methods and technologies enabling molecular investigation at single-cell level. Also, most current methods used to analyze gene expression at RNA level, such as RT-qPCR, do not directly provide quantitative data, but rather comparative ratios between two conditions. A second important need in skin biology is thus to determine the number of RNA molecules in a given cell sample. Here, we describe a workflow that we have set up to meet these specific needs, by means of transcript quantification in cellular micro-samples using flow cytometry sorting and reverse transcription-digital droplet polymerase chain reaction. As a proof-of-principle, the workflow was tested for the detection of transcription factor transcripts expressed at low levels in keratinocyte precursor cells. A linear correlation was found between quantification values and keratinocyte input numbers in a low quantity range from 40 cells to 1 cell. Interpretable signals were repeatedly obtained from single-cell samples corresponding to estimated expression levels as low as 10-20 transcript copies per keratinocyte or less. The present workflow may have broad applications for the detection and quantification of low-abundance nucleic acid species in single cells, opening up perspectives for the study of cell-to-cell genetic and molecular heterogeneity. Interestingly, the process described here does not require internal references such as house-keeping gene expression, as it is initiated with defined cell numbers, precisely sorted by flow cytometry.
Collapse
Affiliation(s)
- Frédéric Auvré
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Fontenay-aux-Roses, France.,Université Paris-Diderot, Paris 7, France.,Université Paris-Saclay, Paris 11, France
| | - Julien Coutier
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Fontenay-aux-Roses, France.,Université Paris-Diderot, Paris 7, France.,Université Paris-Saclay, Paris 11, France
| | - Michèle T Martin
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France.,INSERM U967, Fontenay-aux-Roses, France.,Université Paris-Diderot, Paris 7, France.,Université Paris-Saclay, Paris 11, France
| | - Nicolas O Fortunel
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, CEA/DRF/IBFJ/IRCM, Evry, France. .,INSERM U967, Fontenay-aux-Roses, France. .,Université Paris-Diderot, Paris 7, France. .,Université Paris-Saclay, Paris 11, France.
| |
Collapse
|
20
|
Keyes BE, Fuchs E. Stem cells: Aging and transcriptional fingerprints. J Cell Biol 2017; 217:79-92. [PMID: 29070608 PMCID: PMC5748991 DOI: 10.1083/jcb.201708099] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 12/31/2022] Open
Abstract
Keyes and Fuchs discuss the decline in stem cell renewal and function with aging and the ensuing consequences on tissue homeostasis and regeneration. Stem cells are imbued with unique qualities. They have the capacity to propagate themselves through symmetric divisions and to divide asymmetrically to engender new cells that can progress to differentiate into tissue-specific, terminal cell types. Armed with these qualities, stem cells in adult tissues are tasked with replacing decaying cells and regenerating tissue after injury to maintain optimal tissue function. With increasing age, stem cell functional abilities decline, resulting in reduced organ function and delays in tissue repair. Here, we review the effect of aging in five well-studied adult murine stem cell populations and explore age-related declines in stem cell function and their consequences for stem cell self-renewal, tissue homeostasis, and regeneration. Finally, we examine transcriptional changes that have been documented in aged stem cell populations and discuss new questions and future directions that this collection of data has uncovered.
Collapse
Affiliation(s)
| | - Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY
| |
Collapse
|
21
|
Reinhold D, Morrow JD, Jacobson S, Hu J, Ringel B, Seibold MA, Hersh CP, Kechris KJ, Bowler RP. Meta-analysis of peripheral blood gene expression modules for COPD phenotypes. PLoS One 2017; 12:e0185682. [PMID: 29016655 PMCID: PMC5633174 DOI: 10.1371/journal.pone.0185682] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 09/18/2017] [Indexed: 12/22/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) occurs typically in current or former smokers, but only a minority of people with smoking history develops the disease. Besides environmental factors, genetics is an important risk factor for COPD. However, the relationship between genetics, environment and phenotypes is not well understood. Sample sizes for genome-wide expression studies based on lung tissue have been small due to the invasive nature of sample collection. Increasing evidence for the systemic nature of the disease makes blood a good alternative source to study the disease, but there have also been few large-scale blood genomic studies in COPD. Due to the complexity and heterogeneity of COPD, examining groups of interacting genes may have more relevance than identifying individual genes. Therefore, we used Weighted Gene Co-expression Network Analysis to find groups of genes (modules) that are highly connected. However, module definitions may vary between individual data sets. To alleviate this problem, we used a consensus module definition based on two cohorts, COPDGene and ECLIPSE. We studied the relationship between the consensus modules and COPD phenotypes airflow obstruction and emphysema. We also used these consensus module definitions on an independent cohort (TESRA) and performed a meta analysis involving all data sets. We found several modules that are associated with COPD phenotypes, are enriched in functional categories and are overrepresented for cell-type specific genes. Of the 14 consensus modules, three were strongly associated with airflow obstruction (meta p ≤ 0.0002), and two had some association with emphysema (meta p ≤ 0.06); some associations were stronger in the case-control cohorts, and others in the cases-only subcohorts. Gene Ontology terms that were overrepresented included “immune response” and “defense response.” The cell types whose type-specific genes were overrepresented in modules (p < 0.05) included natural killer cells, dendritic cells, and neutrophils. Together, this is the largest investigation of gene blood expression in COPD with 469 cases in COPDGene, ECLIPSE and TESRA combined, with 6267 genes common to all data sets. Additional, we have 42 and 83 controls in COPDGene and ECLIPSE, respectively.
Collapse
Affiliation(s)
- Dominik Reinhold
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Denver, Aurora, Colorado, United States of America
- * E-mail:
| | - Jarrett D. Morrow
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, United States of America
| | - Sean Jacobson
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Junxiao Hu
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Benjamin Ringel
- Center for Genes, Environment and Health, Department of Pediatrics, National Jewish Health, Denver, Colorado, United States of America
| | - Max A. Seibold
- Center for Genes, Environment and Health, Department of Pediatrics, National Jewish Health, Denver, Colorado, United States of America
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Craig P. Hersh
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, United States of America
| | - Katerina J. Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Russell P. Bowler
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| |
Collapse
|
22
|
Krebsbach PH, Villa-Diaz LG. The Role of Integrin α6 (CD49f) in Stem Cells: More than a Conserved Biomarker. Stem Cells Dev 2017; 26:1090-1099. [PMID: 28494695 DOI: 10.1089/scd.2016.0319] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Stem cells have the capacity for self-renewal and differentiation into specialized cells that form and repopulated all tissues and organs, from conception to adult life. Depending on their capacity for differentiation, stem cells are classified as totipotent (ie, zygote), pluripotent (ie, embryonic stem cells), multipotent (ie, neuronal stem cells, hematopoietic stem cells, epithelial stem cells, etc.), and unipotent (ie, spermatogonial stem cells). Adult or tissue-specific stem cells reside in specific niches located in, or nearby, their organ or tissue of origin. There, they have microenvironmental support to remain quiescent, to proliferate as undifferentiated cells (self-renewal), and to differentiate into progenitors or terminally differentiated cells that migrate from the niche to perform specialized functions. The presence of proteins at the cell surface is often used to identify, classify, and isolate stem cells. Among the diverse groups of cell surface proteins used for these purposes, integrin α6, also known as CD49f, may be the only biomarker commonly found in more than 30 different populations of stem cells, including some cancer stem cells. This broad expression among stem cell populations indicates that integrin α6 may play an important and conserved role in stem cell biology, which is reaffirmed by recent demonstrations of its role maintaining self-renewal of pluripotent stem cells and breast and glioblastoma cancer stem cells. Therefore, this review intends to highlight and synthesize new findings on the importance of integrin α6 in stem cell biology.
Collapse
Affiliation(s)
- Paul H Krebsbach
- 1 School of Dentistry, University of California , Los Angeles, California
| | - Luis G Villa-Diaz
- 2 Department of Biological Sciences, Oakland University , Rochester, Michigan
| |
Collapse
|
23
|
Okawa S, Gagrica S, Blin C, Ender C, Pollard SM, Krijgsveld J. Proteome and Secretome Characterization of Glioblastoma-Derived Neural Stem Cells. Stem Cells 2017; 35:967-980. [PMID: 27870168 PMCID: PMC6135235 DOI: 10.1002/stem.2542] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 10/11/2016] [Indexed: 12/14/2022]
Abstract
Glioblastoma multiforme (GBM) (grade IV astrocytoma) is the most common and aggressive primary brain tumor. GBM consists of heterogeneous cell types including a subset of stem cell-like cells thought to sustain tumor growth. These tumor-initiating glioblastoma multiforme-derived neural stem (GNS) cells as well as their genetically normal neural stem (NS) counterparts can be propagated in culture as relatively pure populations. Here, we perform quantitative proteomics to globally characterize and compare total proteome plus the secreted proteome (secretome) between GNS cells and NS cells. Proteins and pathways that distinguish malignant cancer (GNS) stem cells from their genetically normal counterparts (NS cells) might have value as new biomarkers or therapeutic targets. Our analysis identified and quantified ∼7,500 proteins in the proteome and ∼2,000 in the secretome, 447 and 138 of which were differentially expressed, respectively. Notable tumor-associated processes identified using gene set enrichment analysis included: extracellular matrix interactions, focal adhesion, cell motility, and cell signaling. We focused on differentially expressed surface proteins, and identified 26 that participate in ligand-receptor pairs that play a prominent role in tumorigenesis. Immunocytochemistry and immunoblotting confirmed that CD9, a recently identified marker of adult subventricular zone NS cells, was consistently enriched across a larger set of primary GNS cell lines. CD9 may, therefore, have value as a GNS-specific surface marker and a candidate therapeutic target. Altogether, these findings support the notion that increased cell-matrix and cell-cell adhesion molecules play a crucial role in promoting the tumor initiating and infiltrative properties of GNS cells. Stem Cells 2017;35:967-980.
Collapse
Affiliation(s)
- Satoshi Okawa
- European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, Heidelberg 69117, Germany
| | - Sladjana Gagrica
- Department of Cancer Biology, Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London, WC1E 6BT, UK
| | - Carla Blin
- Department of Cancer Biology, Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London, WC1E 6BT, UK
| | - Christine Ender
- Department of Cancer Biology, Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London, WC1E 6BT, UK
| | - Steven M. Pollard
- Department of Cancer Biology, Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London, WC1E 6BT, UK
| | - Jeroen Krijgsveld
- European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, Heidelberg 69117, Germany
- German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany
- CellNetworks - Cluster of Excellence, and Heidelberg University, Heidelberg, Germany
| |
Collapse
|
24
|
Grzybek M, Golonko A, Walczak M, Lisowski P. Epigenetics of cell fate reprogramming and its implications for neurological disorders modelling. Neurobiol Dis 2016; 99:84-120. [PMID: 27890672 DOI: 10.1016/j.nbd.2016.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 11/03/2016] [Accepted: 11/21/2016] [Indexed: 02/06/2023] Open
Abstract
The reprogramming of human induced pluripotent stem cells (hiPSCs) proceeds in a stepwise manner with reprogramming factors binding and epigenetic composition changes during transition to maintain the epigenetic landscape, important for pluripotency. There arises a question as to whether the aberrant epigenetic state after reprogramming leads to epigenetic defects in induced stem cells causing unpredictable long term effects in differentiated cells. In this review, we present a comprehensive view of epigenetic alterations accompanying reprogramming, cell maintenance and differentiation as factors that influence applications of hiPSCs in stem cell based technologies. We conclude that sample heterogeneity masks DNA methylation signatures in subpopulations of cells and thus believe that beside a genetic evaluation, extensive epigenomic screening should become a standard procedure to ensure hiPSCs state before they are used for genome editing and differentiation into neurons of interest. In particular, we suggest that exploitation of the single-cell composition of the epigenome will provide important insights into heterogeneity within hiPSCs subpopulations to fast forward development of reliable hiPSC-based analytical platforms in neurological disorders modelling and before completed hiPSC technology will be implemented in clinical approaches.
Collapse
Affiliation(s)
- Maciej Grzybek
- Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 12, 20-950 Lublin, Poland; Department of Molecular Biology, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzębiec, Postępu 36A, 05-552 Magdalenka, Poland.
| | - Aleksandra Golonko
- Department of Biotechnology, Faculty of Civil and Environmental Engineering, Bialystok University of Technology, Wiejska 45E, 15-351 Bialystok, Poland.
| | - Marta Walczak
- Department of Animal Behavior, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzębiec, Postępu 36A, 05-552 Magdalenka, Poland.
| | - Pawel Lisowski
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzębiec, Postępu 36A, 05-552 Magdalenka, Poland; iPS Cell-Based Disease Modelling Group, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Robert-Rössle-Str. 10, 13092 Berlin, Germany.
| |
Collapse
|
25
|
Bhartiya D, Shaikh A, Anand S, Patel H, Kapoor S, Sriraman K, Parte S, Unni S. Endogenous, very small embryonic-like stem cells: critical review, therapeutic potential and a look ahead. Hum Reprod Update 2016; 23:41-76. [PMID: 27614362 DOI: 10.1093/humupd/dmw030] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/27/2016] [Accepted: 08/04/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Both pluripotent very small embryonic-like stem cells (VSELs) and induced pluripotent stem (iPS) cells were reported in 2006. In 2012, a Nobel Prize was awarded for iPS technology whereas even today the very existence of VSELs is not well accepted. The underlying reason is that VSELs exist in low numbers, remain dormant under homeostatic conditions, are very small in size and do not pellet down at 250-280g. The VSELs maintain life-long tissue homeostasis, serve as a backup pool for adult stem cells and are mobilized under stress conditions. An imbalance in VSELs function (uncontrolled proliferation) may result in cancer. SEARCH METHODS The electronic database 'Medline/Pubmed' was systematically searched with the subject heading term 'very small embryonic-like stem cells'. OBJECTIVE AND RATIONALE The most primitive stem cells that undergo asymmetric cell divisions to self-renew and give rise to progenitors still remain elusive in the hematopoietic system and testes, while the presence of stem cells in ovary is still being debated. We propose to review the available literature on VSELs, the methods of their isolation and characterization, their ontogeny, how they compare with embryonic stem (ES) cells, primordial germ cells (PGCs) and iPS cells, and their role in maintaining tissue homeostasis. The review includes a look ahead on how VSELs will result in paradigm shifts in basic reproductive biology. OUTCOMES Adult tissue-specific stem cells including hematopoietic, spermatogonial, ovarian and mesenchymal stem cells have good proliferation potential and are indeed committed progenitors (with cytoplasmic OCT-4), which arise by asymmetric cell divisions of pluripotent VSELs (with nuclear OCT-4). VSELs are the most primitive stem cells and postulated to be an overlapping population with the PGCs. Rather than migrating only to the gonads, PGCs migrate and survive in various adult body organs throughout life as VSELs. VSELs express both pluripotent and PGC-specific markers and are epigenetically and developmentally more mature compared with ES cells obtained from the inner cell mass of a blastocyst-stage embryo. As a result, VSELs readily differentiate into three embryonic germ layers and spontaneously give rise to both sperm and oocytes in vitro. Like PGCs, VSELs do not divide readily in culture, nor produce teratoma or integrate in the developing embryo. But this property of being relatively quiescent allows endogenous VSELs to survive various kinds of toxic insults. VSELs that survive oncotherapy can be targeted to induce endogenous regeneration of non-functional gonads. Transplanting healthy niche (mesenchymal) cells have resulted in improved gonadal function and live births. WIDER IMPLICATIONS Being quiescent, VSELs possibly do not accumulate genomic (nuclear or mitochondrial) mutations and thus may be ideal endogenous, pluripotent stem cell candidates for regenerative and reproductive medicine. The presence of VSELs in adult gonads and the fact that they survive oncotherapy may obviate the need to bank gonadal tissue for fertility preservation prior to oncotherapy. VSELs and their ability to undergo spermatogenesis/neo-oogenesis in the presence of a healthy niche will help identify newer strategies toward fertility restoration in cancer survivors, delaying menopause and also enabling aged mothers to have better quality eggs.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Ambreen Shaikh
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Sandhya Anand
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Hiren Patel
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Sona Kapoor
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Kalpana Sriraman
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India.,The Foundation for Medical Research, 84-A, RG Thadani Marg, Worli, Mumbai 400018, India
| | - Seema Parte
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India.,Department of Physiology, James Graham Brown Cancer Centre, University of Louisville School of Medicine, 2301 S 3rd St, Louisville, KY 40202, USA
| | - Sreepoorna Unni
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India.,Inter Disciplinary Studies Department, University College, Zayed University, Academic City, PO Box 19282, Dubai, United Arab Emirates
| |
Collapse
|
26
|
Niwa O, Barcellos-Hoff MH, Globus RK, Harrison JD, Hendry JH, Jacob P, Martin MT, Seed TM, Shay JW, Story MD, Suzuki K, Yamashita S. ICRP Publication 131: Stem Cell Biology with Respect to Carcinogenesis Aspects of Radiological Protection. Ann ICRP 2016; 44:7-357. [PMID: 26637346 DOI: 10.1177/0146645315595585] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This report provides a review of stem cells/progenitor cells and their responses to ionising radiation in relation to issues relevant to stochastic effects of radiation that form a major part of the International Commission on Radiological Protection's system of radiological protection. Current information on stem cell characteristics, maintenance and renewal, evolution with age, location in stem cell 'niches', and radiosensitivity to acute and protracted exposures is presented in a series of substantial reviews as annexes concerning haematopoietic tissue, mammary gland, thyroid, digestive tract, lung, skin, and bone. This foundation of knowledge of stem cells is used in the main text of the report to provide a biological insight into issues such as the linear-no-threshold (LNT) model, cancer risk among tissues, dose-rate effects, and changes in the risk of radiation carcinogenesis by age at exposure and attained age. Knowledge of the biology and associated radiation biology of stem cells and progenitor cells is more developed in tissues that renew fairly rapidly, such as haematopoietic tissue, intestinal mucosa, and epidermis, although all the tissues considered here possess stem cell populations. Important features of stem cell maintenance, renewal, and response are the microenvironmental signals operating in the niche residence, for which a well-defined spatial location has been identified in some tissues. The identity of the target cell for carcinogenesis continues to point to the more primitive stem cell population that is mostly quiescent, and hence able to accumulate the protracted sequence of mutations necessary to result in malignancy. In addition, there is some potential for daughter progenitor cells to be target cells in particular cases, such as in haematopoietic tissue and in skin. Several biological processes could contribute to protecting stem cells from mutation accumulation: (a) accurate DNA repair; (b) rapidly induced death of injured stem cells; (c) retention of the DNA parental template strand during divisions in some tissue systems, so that mutations are passed to the daughter differentiating cells and not retained in the parental cell; and (d) stem cell competition, whereby undamaged stem cells outcompete damaged stem cells for residence in the niche. DNA repair mainly occurs within a few days of irradiation, while stem cell competition requires weeks or many months depending on the tissue type. The aforementioned processes may contribute to the differences in carcinogenic radiation risk values between tissues, and may help to explain why a rapidly replicating tissue such as small intestine is less prone to such risk. The processes also provide a mechanistic insight relevant to the LNT model, and the relative and absolute risk models. The radiobiological knowledge also provides a scientific insight into discussions of the dose and dose-rate effectiveness factor currently used in radiological protection guidelines. In addition, the biological information contributes potential reasons for the age-dependent sensitivity to radiation carcinogenesis, including the effects of in-utero exposure.
Collapse
|
27
|
Martin MT, Vulin A, Hendry JH. Human epidermal stem cells: Role in adverse skin reactions and carcinogenesis from radiation. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 770:349-368. [PMID: 27919341 DOI: 10.1016/j.mrrev.2016.08.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/12/2016] [Accepted: 08/13/2016] [Indexed: 02/06/2023]
Abstract
In human skin, keratinopoiesis is based on a functional hierarchy among keratinocytes, with rare slow-cycling stem cells responsible for the long-term maintenance of the tissue through their self-renewal potential, and more differentiated daughter progenitor cells actively cycling to permit epidermal renewal and turn-over every month. Skin is a radio-responsive tissue, developing all types of radiation damage and pathologies, including early tissue reactions such as dysplasia and denudation in epidermis, and later fibrosis in the dermis and acanthosis in epidermis, with the TGF-beta 1 pathway as a known master switch. Also there is a risk of basal cell carcinoma, which arises from epidermal keratinocytes, notably after oncogenic events in PTCH1 or TP53 genes. This review will cover the mechanisms of adverse human skin reactions and carcinogenesis after various types of exposures to ionizing radiation, with comparison with animal data when necessary, and will discuss the possible role of stem cells and their progeny in the development of these disorders. The main endpoints presented are basal cell intrinsic radiosensitivity, genomic stability, individual factors of risk, dose specific responses, major molecular pathways involved and the cellular origin of skin reactions and cancer. Although major advances have been obtained in recent years, the precise implications of epidermal stem cells and their progeny in these processes are not yet fully characterized.
Collapse
Affiliation(s)
- Michèle T Martin
- CEA/DRF/IRCM/LGRK, 91057 Evry, France; INSERM U967, 92265 Fontenay aux Roses, Cedex, France; Université Paris-Diderot, Paris 7, France; Université Paris-Saclay, Paris 11, France.
| | - Adeline Vulin
- CEA/DRF/IRCM/LGRK, 91057 Evry, France; INSERM U967, 92265 Fontenay aux Roses, Cedex, France; Université Paris-Diderot, Paris 7, France; Université Paris-Saclay, Paris 11, France
| | - Jolyon H Hendry
- Christie Medical Physics and Engineering, Christie Hospital and University of Manchester, Manchester, United Kingdom
| |
Collapse
|
28
|
Current Stem Cell Biomarkers and Their Functional Mechanisms in Prostate Cancer. Int J Mol Sci 2016; 17:ijms17071163. [PMID: 27447616 PMCID: PMC4964535 DOI: 10.3390/ijms17071163] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/07/2016] [Accepted: 07/09/2016] [Indexed: 12/12/2022] Open
Abstract
Currently there is little effective treatment available for castration resistant prostate cancer, which is responsible for the majority of prostate cancer related deaths. Emerging evidence suggested that cancer stem cells might play an important role in resistance to traditional cancer therapies, and the studies of cancer stem cells (including specific isolation and targeting on those cells) might benefit the discovery of novel treatment of prostate cancer, especially castration resistant disease. In this review, we summarized major biomarkers for prostate cancer stem cells, as well as their functional mechanisms and potential application in clinical diagnosis and treatment of patients.
Collapse
|
29
|
Semantic Signature: Comparative Interpretation of Gene Expression on a Semantic Space. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2016; 2016:5174503. [PMID: 27242916 PMCID: PMC4868886 DOI: 10.1155/2016/5174503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/23/2016] [Indexed: 11/17/2022]
Abstract
Background. Interpretation of microarray data remains challenging because biological meaning should be extracted from enormous numeric matrices and be presented explicitly. Moreover, huge public repositories of microarray dataset are ready to be exploited for comparative analysis. This study aimed to provide a platform where essential implication of a microarray experiment could be visually expressed and various microarray datasets could be intuitively compared. Results. On the semantic space, gene sets from Molecular Signature Database (MSigDB) were plotted as landmarks and their relative distances were calculated by Lin's semantic similarity measure. By formal concept analysis, a microarray dataset was transformed into a concept lattice with gene clusters as objects and Gene Ontology terms as attributes. Concepts of a lattice were located on the semantic space reflecting semantic distance from landmarks and edges between concepts were drawn; consequently, a specific geographic pattern could be observed from a microarray dataset. We termed a distinctive geography shared by microarray datasets of the same category as “semantic signature.” Conclusions. “Semantic space,” a map of biological entities, could serve as a universal platform for comparative microarray analysis. When microarray data were displayed on the semantic space as concept lattices, “semantic signature,” characteristic geography for a microarray experiment, could be discovered.
Collapse
|
30
|
Glioblastoma Stem Cells Microenvironment: The Paracrine Roles of the Niche in Drug and Radioresistance. Stem Cells Int 2016; 2016:6809105. [PMID: 26880981 PMCID: PMC4736577 DOI: 10.1155/2016/6809105] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/09/2015] [Accepted: 11/10/2015] [Indexed: 12/13/2022] Open
Abstract
Among all solid tumors, the high-grade glioma appears to be the most vascularized one. In fact, "microvascular hyperplasia" is a hallmark of GBM. An altered vascular network determines irregular blood flow, so that tumor cells spread rapidly beyond the diffusion distance of oxygen in the tissue, with the consequent formation of hypoxic or anoxic areas, where the bulk of glioblastoma stem cells (GSCs) reside. The response to this event is the induction of angiogenesis, a process mediated by hypoxia inducible factors. However, this new capillary network is not efficient in maintaining a proper oxygen supply to the tumor mass, thereby causing an oxygen gradient within the neoplastic zone. This microenvironment helps GSCs to remain in a "quiescent" state preserving their potential to proliferate and differentiate, thus protecting them by the effects of chemo- and radiotherapy. Recent evidences suggest that responses of glioblastoma to standard therapies are determined by the microenvironment of the niche, where the GSCs reside, allowing a variety of mechanisms that contribute to the chemo- and radioresistance, by preserving GSCs. It is, therefore, crucial to investigate the components/factors of the niche in order to formulate new adjuvant therapies rendering more efficiently the gold standard therapies for this neoplasm.
Collapse
|
31
|
Yard B, Chie EK, Adams DJ, Peacock C, Abazeed ME. Radiotherapy in the Era of Precision Medicine. Semin Radiat Oncol 2015; 25:227-36. [DOI: 10.1016/j.semradonc.2015.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
32
|
Novianti PW, van der Tweel I, Jong VL, Roes KC, Eijkemans MJ. An Application of Sequential Meta-Analysis to Gene Expression Studies. Cancer Inform 2015; 14:1-10. [PMID: 26401096 PMCID: PMC4567049 DOI: 10.4137/cin.s27718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/03/2015] [Accepted: 06/04/2015] [Indexed: 11/15/2022] Open
Abstract
Most of the discoveries from gene expression data are driven by a study claiming an optimal subset of genes that play a key role in a specific disease. Meta-analysis of the available datasets can help in getting concordant results so that a real-life application may be more successful. Sequential meta-analysis (SMA) is an approach for combining studies in chronological order while preserving the type I error and pre-specifying the statistical power to detect a given effect size. We focus on the application of SMA to find gene expression signatures across experiments in acute myeloid leukemia. SMA of seven raw datasets is used to evaluate whether the accumulated samples show enough evidence or more experiments should be initiated. We found 313 differentially expressed genes, based on the cumulative information of the experiments. SMA offers an alternative to existing methods in generating a gene list by evaluating the adequacy of the cumulative information.
Collapse
Affiliation(s)
- Putri W Novianti
- Biostatistics and Research Support, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ingeborg van der Tweel
- Biostatistics and Research Support, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Victor L Jong
- Biostatistics and Research Support, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands ; Department of Viroscience, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Kit Cb Roes
- Biostatistics and Research Support, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marinus Jc Eijkemans
- Biostatistics and Research Support, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
33
|
Larsimont JC, Youssef KK, Sánchez-Danés A, Sukumaran V, Defrance M, Delatte B, Liagre M, Baatsen P, Marine JC, Lippens S, Guerin C, Del Marmol V, Vanderwinden JM, Fuks F, Blanpain C. Sox9 Controls Self-Renewal of Oncogene Targeted Cells and Links Tumor Initiation and Invasion. Cell Stem Cell 2015; 17:60-73. [PMID: 26095047 DOI: 10.1016/j.stem.2015.05.008] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 03/30/2015] [Accepted: 05/15/2015] [Indexed: 01/03/2023]
Abstract
Sox9 is a transcription factor expressed in most solid tumors. However, the molecular mechanisms underlying Sox9 function during tumorigenesis remain unclear. Here, using a genetic mouse model of basal cell carcinoma (BCC), the most frequent cancer in humans, we show that Sox9 is expressed from the earliest step of tumor formation in a Wnt/β-catenin-dependent manner. Deletion of Sox9 together with the constitutive activation of Hedgehog signaling completely prevents BCC formation and leads to a progressive loss of oncogene-expressing cells. Transcriptional profiling of oncogene-expressing cells with Sox9 deletion, combined with in vivo ChIP sequencing, uncovers a cancer-specific gene network regulated by Sox9 that promotes stemness, extracellular matrix deposition, and cytoskeleton remodeling while repressing epidermal differentiation. Our study identifies the molecular mechanisms regulated by Sox9 that link tumor initiation and invasion.
Collapse
Affiliation(s)
| | | | | | | | - Matthieu Defrance
- Laboratory of Cancer Epigenetics, Faculty of Medicine, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Benjamin Delatte
- Laboratory of Cancer Epigenetics, Faculty of Medicine, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Mélanie Liagre
- Université Libre de Bruxelles, IRIBHM, Brussels 1070, Belgium
| | - Pieter Baatsen
- EM-Facility EMoNe, VIB BIO Imaging Core, Center for Human Genetics Katholieke Universiteit Leuven, Leuven 3000, Belgium
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for the Biology of Disease, VIB, Leuven 3000, Belgium
| | - Saskia Lippens
- Inflammation Research Center, Image Core Facility, VIB, Ghent 9052, Belgium; VIB Bio Imaging Core, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium
| | - Christopher Guerin
- Inflammation Research Center, Image Core Facility, VIB, Ghent 9052, Belgium; VIB Bio Imaging Core, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium
| | - Véronique Del Marmol
- Department of Dermatology, Erasme Hospital, Université Libre de Bruxelles, Brussels 1070, Belgium
| | | | - Francois Fuks
- Laboratory of Cancer Epigenetics, Faculty of Medicine, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Cédric Blanpain
- Université Libre de Bruxelles, IRIBHM, Brussels 1070, Belgium; WELBIO, Université Libre de Bruxelles, Brussels 1070, Belgium.
| |
Collapse
|
34
|
Pinto JP, Kalathur RK, Oliveira DV, Barata T, Machado RSR, Machado S, Pacheco-Leyva I, Duarte I, Futschik ME. StemChecker: a web-based tool to discover and explore stemness signatures in gene sets. Nucleic Acids Res 2015; 43:W72-7. [PMID: 26007653 PMCID: PMC4489266 DOI: 10.1093/nar/gkv529] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/10/2015] [Indexed: 11/25/2022] Open
Abstract
Stem cells present unique regenerative abilities, offering great potential for treatment of prevalent pathologies such as diabetes, neurodegenerative and heart diseases. Various research groups dedicated significant effort to identify sets of genes—so-called stemness signatures—considered essential to define stem cells. However, their usage has been hindered by the lack of comprehensive resources and easy-to-use tools. For this we developed StemChecker, a novel stemness analysis tool, based on the curation of nearly fifty published stemness signatures defined by gene expression, RNAi screens, Transcription Factor (TF) binding sites, literature reviews and computational approaches. StemChecker allows researchers to explore the presence of stemness signatures in user-defined gene sets, without carrying-out lengthy literature curation or data processing. To assist in exploring underlying regulatory mechanisms, we collected over 80 target gene sets of TFs associated with pluri- or multipotency. StemChecker presents an intuitive graphical display, as well as detailed statistical results in table format, which helps revealing transcriptionally regulatory programs, indicating the putative involvement of stemness-associated processes in diseases like cancer. Overall, StemChecker substantially expands the available repertoire of online tools, designed to assist the stem cell biology, developmental biology, regenerative medicine and human disease research community. StemChecker is freely accessible at http://stemchecker.sysbiolab.eu.
Collapse
Affiliation(s)
- José P Pinto
- Systems Biology and Bioinformatics Laboratory (SysBioLab), University of Algarve, Faro, Algarve, 8005-139, Portugal
| | - Ravi K Kalathur
- Systems Biology and Bioinformatics Laboratory (SysBioLab), University of Algarve, Faro, Algarve, 8005-139, Portugal Experimental and Clinical Cell Therapy Institute, Spinal Cord and Tissue Regeneration Center Salzburg, Paracelsus Medizinische Privatuniversität, Salzburg, A-5020, Austria
| | - Daniel V Oliveira
- Systems Biology and Bioinformatics Laboratory (SysBioLab), University of Algarve, Faro, Algarve, 8005-139, Portugal
| | - Tânia Barata
- Systems Biology and Bioinformatics Laboratory (SysBioLab), University of Algarve, Faro, Algarve, 8005-139, Portugal
| | - Rui S R Machado
- Systems Biology and Bioinformatics Laboratory (SysBioLab), University of Algarve, Faro, Algarve, 8005-139, Portugal
| | - Susana Machado
- Systems Biology and Bioinformatics Laboratory (SysBioLab), University of Algarve, Faro, Algarve, 8005-139, Portugal
| | - Ivette Pacheco-Leyva
- Systems Biology and Bioinformatics Laboratory (SysBioLab), University of Algarve, Faro, Algarve, 8005-139, Portugal
| | - Isabel Duarte
- Systems Biology and Bioinformatics Laboratory (SysBioLab), University of Algarve, Faro, Algarve, 8005-139, Portugal
| | - Matthias E Futschik
- Systems Biology and Bioinformatics Laboratory (SysBioLab), University of Algarve, Faro, Algarve, 8005-139, Portugal Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Algarve, 8005-139, Portugal
| |
Collapse
|
35
|
Method designed to respect molecular heterogeneity can profoundly correct present data interpretations for genome-wide expression analysis. PLoS One 2015; 10:e0121154. [PMID: 25793610 PMCID: PMC4368820 DOI: 10.1371/journal.pone.0121154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/20/2015] [Indexed: 11/27/2022] Open
Abstract
Although genome-wide expression analysis has become a routine tool for gaining insight into molecular mechanisms, extraction of information remains a major challenge. It has been unclear why standard statistical methods, such as the t-test and ANOVA, often lead to low levels of reproducibility, how likely applying fold-change cutoffs to enhance reproducibility is to miss key signals, and how adversely using such methods has affected data interpretations. We broadly examined expression data to investigate the reproducibility problem and discovered that molecular heterogeneity, a biological property of genetically different samples, has been improperly handled by the statistical methods. Here we give a mathematical description of the discovery and report the development of a statistical method, named HTA, for better handling molecular heterogeneity. We broadly demonstrate the improved sensitivity and specificity of HTA over the conventional methods and show that using fold-change cutoffs has lost much information. We illustrate the especial usefulness of HTA for heterogeneous diseases, by applying it to existing data sets of schizophrenia, bipolar disorder and Parkinson’s disease, and show it can abundantly and reproducibly uncover disease signatures not previously detectable. Based on 156 biological data sets, we estimate that the methodological issue has affected over 96% of expression studies and that HTA can profoundly correct 86% of the affected data interpretations. The methodological advancement can better facilitate systems understandings of biological processes, render biological inferences that are more reliable than they have hitherto been and engender translational medical applications, such as identifying diagnostic biomarkers and drug prediction, which are more robust.
Collapse
|
36
|
Braun E. The unforeseen challenge: from genotype-to-phenotype in cell populations. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2015; 78:036602. [PMID: 25719211 DOI: 10.1088/0034-4885/78/3/036602] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Biological cells present a paradox, in that they show simultaneous stability and flexibility, allowing them to adapt to new environments and to evolve over time. The emergence of stable cell states depends on genotype-to-phenotype associations, which essentially reflect the organization of gene regulatory modes. The view taken here is that cell-state organization is a dynamical process in which the molecular disorder manifests itself in a macroscopic order. The genome does not determine the ordered cell state; rather, it participates in this process by providing a set of constraints on the spectrum of regulatory modes, analogous to boundary conditions in physical dynamical systems. We have developed an experimental framework, in which cell populations are exposed to unforeseen challenges; novel perturbations they had not encountered before along their evolutionary history. This approach allows an unbiased view of cell dynamics, uncovering the potential of cells to evolve and develop adapted stable states. In the last decade, our experiments have revealed a coherent set of observations within this framework, painting a picture of the living cell that in many ways is not aligned with the conventional one. Of particular importance here, is our finding that adaptation of cell-state organization is essentially an efficient exploratory dynamical process rather than one founded on random mutations. Based on our framework, a set of concepts underlying cell-state organization-exploration evolving by global, non-specific, dynamics of gene activity-is presented here. These concepts have significant consequences for our understanding of the emergence and stabilization of a cell phenotype in diverse biological contexts. Their implications are discussed for three major areas of biological inquiry: evolution, cell differentiation and cancer. There is currently no unified theoretical framework encompassing the emergence of order, a stable state, in the living cell. Hopefully, the integrated picture described here will provide a modest contribution towards a physics theory of the cell.
Collapse
Affiliation(s)
- Erez Braun
- Department of Physics and Network Biology Research Laboratories, Technion, Haifa 32000, Israel
| |
Collapse
|
37
|
Huh YH, Noh M, Burden FR, Chen JC, Winkler DA, Sherley JL. Sparse feature selection identifies H2A.Z as a novel, pattern-specific biomarker for asymmetrically self-renewing distributed stem cells. Stem Cell Res 2015; 14:144-54. [PMID: 25636161 DOI: 10.1016/j.scr.2014.12.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 12/19/2014] [Accepted: 12/26/2014] [Indexed: 01/17/2023] Open
Abstract
There is a long-standing unmet clinical need for biomarkers with high specificity for distributed stem cells (DSCs) in tissues, or for use in diagnostic and therapeutic cell preparations (e.g., bone marrow). Although DSCs are essential for tissue maintenance and repair, accurate determination of their numbers for medical applications has been problematic. Previous searches for biomarkers expressed specifically in DSCs were hampered by difficulty obtaining pure DSCs and by the challenges in mining complex molecular expression data. To identify such useful and specific DSC biomarkers, we combined a novel sparse feature selection method with combinatorial molecular expression data focused on asymmetric self-renewal, a conspicuous property of DSCs. The analysis identified reduced expression of the histone H2A variant H2A.Z as a superior molecular discriminator for DSC asymmetric self-renewal. Subsequent molecular expression studies showed H2A.Z to be a novel "pattern-specific biomarker" for asymmetrically self-renewing cells, with sufficient specificity to count asymmetrically self-renewing DSCs in vitro and potentially in situ.
Collapse
Affiliation(s)
- Yang Hoon Huh
- Division of Electron Microscopic Research, Korea Basic Science Institute, 169-148 Gwahak-ro, Yuseong-gu, Daejeon 305-806, Republic of Korea
| | - Minsoo Noh
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | | | - Jennifer C Chen
- The Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - David A Winkler
- CSIRO Manufacturing Flagship, Clayton, Australia; Monash Institute of Pharmaceutical Sciences, Parkville, Australia; La Trobe Institute for Molecular Science, Bundoora, Australia.
| | | |
Collapse
|
38
|
Trépant AL, Bouchart C, Rorive S, Sauvage S, Decaestecker C, Demetter P, Salmon I. Identification of OLIG2 as the most specific glioblastoma stem cell marker starting from comparative analysis of data from similar DNA chip microarray platforms. Tumour Biol 2014; 36:1943-53. [PMID: 25384509 DOI: 10.1007/s13277-014-2800-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 11/03/2014] [Indexed: 12/15/2022] Open
Abstract
Despite advances in surgical and adjuvant treatments, overall survival of glioblastoma (GBM) patients remains poor. The cancer stem cell concept suggests that a rare stem cell population, called glioma stem cells (GSCs), has high ability to self-renewal leading to recurrence in GBM. The identification of specific markers of GSCs would provide a powerful tool to detect and to characterise them in order to develop targeted therapies. We carried out a comparative analysis based on the identification of inter-study concordances to identify the genes that exhibit at best differential levels of expression between GSC-enriched cell cultures and differentiated tumour cell cultures from independent studies using DNA chip microarray technologies. We finally studied the protein expression of the marker we considered the most specific by immunohistochemistry and semi-quantitative analysis on a retrospective series of 18 GBMs. Of the selected studies, 32 genes were retained. Among them, eight genes were identified to be overexpressed in GSC-enriched cultures compared to differentiated tumour cell cultures. Finally, among the eight genes, oligodendrocyte lineage transcription factor 2 (OLIG2) was characterised by the most different expression level in the "GSC model" compared to the "differentiated tumour cells model". Our approach suggests that OLIG2 is the most specific GSC marker; additional investigations with careful considerations about methodology and strategies of validation are, however, mandatory.
Collapse
Affiliation(s)
- Anne-Laure Trépant
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
39
|
Castellani G, Intrator N, Remondini D. Systems biology and brain activity in neuronal pathways by smart device and advanced signal processing. Front Genet 2014; 5:253. [PMID: 25206359 PMCID: PMC4143968 DOI: 10.3389/fgene.2014.00253] [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: 04/03/2014] [Accepted: 07/10/2014] [Indexed: 01/30/2023] Open
Abstract
Contemporary biomedicine is producing large amount of data, especially within the fields of “omic” sciences. Nevertheless, other fields, such as neuroscience, are producing similar amount of data by using non-invasive techniques such as imaging, functional magnetic resonance and electroencephalography. Nowadays a big challenge and a new research horizon for Systems Biology is to develop methods to integrate and model this data in an unifying framework capable to disentangle this amazing complexity. In this paper we show how methods from genomic data analysis can be applied to brain data. In particular the concept of pathways, networks and multiplex are discussed. These methods can lead to a clear distinction of various regimes of brain activity. Moreover, this method could be the basis for a Systems Biology analysis of brain data and for the integration of these data in a multivariate and multidimensional framework. The feasibility of this integration is strongly dependent from the feature extraction method used. In our case we used an “alphabet” derived from a multi-resolution analysis that is capable to capture the most relevant information from these complex signals.
Collapse
Affiliation(s)
- Gastone Castellani
- Department of Physics and Astronomy, L. Galvani Center for Biocomplexity, Biophysics and Systems Biology, University of Bologna Bologna, Italy
| | - Nathan Intrator
- Department of Computer Science, Exact Sciences Faculty, Tel Aviv University Tel Aviv, Israel
| | - Daniel Remondini
- Department of Physics and Astronomy, L. Galvani Center for Biocomplexity, Biophysics and Systems Biology, University of Bologna Bologna, Italy
| |
Collapse
|
40
|
Pinto JP, Reddy Kalathur RK, Machado RSR, Xavier JM, Bragança J, Futschik ME. StemCellNet: an interactive platform for network-oriented investigations in stem cell biology. Nucleic Acids Res 2014; 42:W154-W160. [PMID: 24852251 PMCID: PMC4086070 DOI: 10.1093/nar/gku455] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 05/02/2014] [Accepted: 05/08/2014] [Indexed: 12/11/2022] Open
Abstract
Stem cells are characterized by their potential for self-renewal and their capacity to differentiate into mature cells. These two key features emerge through the interplay of various factors within complex molecular networks. To provide researchers with a dedicated tool to investigate these networks, we have developed StemCellNet, a versatile web server for interactive network analysis and visualization. It rapidly generates focused networks based on a large collection of physical and regulatory interactions identified in human and murine stem cells. The StemCellNet web-interface has various easy-to-use tools for selection and prioritization of network components, as well as for integration of expression data provided by the user. As a unique feature, the networks generated can be screened against a compendium of stemness-associated genes. StemCellNet can also indicate novel candidate genes by evaluating their connectivity patterns. Finally, an optional dataset of generic interactions, which provides large coverage of the human and mouse proteome, extends the versatility of StemCellNet to other biomedical research areas in which stem cells play important roles, such as in degenerative diseases or cancer. The StemCellNet web server is freely accessible at http://stemcellnet.sysbiolab.eu.
Collapse
Affiliation(s)
- José P Pinto
- Centre for Molecular and Structural Biomedicine, CBME/IBB, LA, University of Algarve, Faro, Algarve 8005-139, Portugal
| | - Ravi Kiran Reddy Kalathur
- Centre for Molecular and Structural Biomedicine, CBME/IBB, LA, University of Algarve, Faro, Algarve 8005-139, Portugal
| | - Rui S R Machado
- Centre for Molecular and Structural Biomedicine, CBME/IBB, LA, University of Algarve, Faro, Algarve 8005-139, Portugal
| | - Joana M Xavier
- Centre for Molecular and Structural Biomedicine, CBME/IBB, LA, University of Algarve, Faro, Algarve 8005-139, Portugal
| | - José Bragança
- Centre for Molecular and Structural Biomedicine, CBME/IBB, LA, University of Algarve, Faro, Algarve 8005-139, Portugal Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Algarve 8005-139, Portugal
| | - Matthias E Futschik
- Centre for Molecular and Structural Biomedicine, CBME/IBB, LA, University of Algarve, Faro, Algarve 8005-139, Portugal Centre of Marine Science, University of Algarve, Faro, Algarve 8005-139, Portugal
| |
Collapse
|
41
|
Hart ML, Neumayer KMH, Vaegler M, Daum L, Amend B, Sievert KD, Di Giovanni S, Kraushaar U, Guenther E, Stenzl A, Aicher WK. Cell-based therapy for the deficient urinary sphincter. Curr Urol Rep 2014; 14:476-87. [PMID: 23824516 DOI: 10.1007/s11934-013-0352-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
When sterile culture techniques of mammalian cells first became state of the art, there was tremendous anticipation that such cells could be eventually applied for therapeutic purposes. The discovery of adult human stem or progenitor cells further motivated scientists to pursue research in cell-based therapies. Although evidence from animal studies suggests that application of cells yields measurable benefits, in urology and many other disciplines, progenitor-cell-based therapies are not yet routinely clinically available. Stress urinary incontinence (SUI) is a condition affecting a large number of patients. The etiology of SUI includes, but is not limited to, degeneration of the urinary sphincter muscle tissue and loss of innervation, as well as anatomical and biomechanical causes. Therefore, different regimens were developed to treat SUI. However, at present, a curative functional treatment is not at hand. A progenitor-cell-based therapy that can tackle the etiology of incontinence, rather than the consequences, is a promising strategy. Therefore, several research teams have intensified their efforts to develop such a therapy for incontinence. Here, we introduce candidate stem and progenitor cells suitable for SUI treatment, show how the functional homogeneity and state of maturity of differentiated cells crucial for proper tissue integration can be assessed electrophysiologically prior to their clinical application, and discuss the trophic potential of adult mesenchymal stromal (or stem) cells in regeneration of neuronal function.
Collapse
Affiliation(s)
- Melanie L Hart
- KFO273, Department of Urology, UKT, University of Tuebingen, Paul-Ehrlich-Str. 15, 72076, Tuebingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Cox-Limpens KEM, Gavilanes AWD, Zimmermann LJI, Vles JSH. Endogenous brain protection: what the cerebral transcriptome teaches us. Brain Res 2014; 1564:85-100. [PMID: 24713346 DOI: 10.1016/j.brainres.2014.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 03/23/2014] [Accepted: 04/01/2014] [Indexed: 02/04/2023]
Abstract
Despite efforts to reduce mortality caused by stroke and perinatal asphyxia, these are still the 2nd largest cause of death worldwide in the age groups they affect. Furthermore, survivors of cerebral hypoxia-ischemia often suffer neurological morbidities. A better understanding of pathophysiological mechanisms in focal and global brain ischemia will contribute to the development of tailored therapeutic strategies. Similarly, insight into molecular pathways involved in preconditioning-induced brain protection will provide possibilities for future treatment. Microarray technology is a great tool for investigating large scale gene expression, and has been used in many experimental studies of cerebral ischemia and preconditioning to unravel molecular (patho-) physiology. However, the amount of data across microarray studies can be daunting and hard to interpret which is why we aim to provide a clear overview of available data in experimental rodent models. Findings for both injurious ischemia and preconditioning are reviewed under separate subtopics such as cellular stress, inflammation, cytoskeleton and cell signaling. Finally, we investigated the transcriptome signature of brain protection across preconditioning studies in search of transcripts that were expressed similarly across studies. Strikingly, when comparing genes discovered by single-gene analysis we observed only 15 genes present in two studies or more. We subjected these 15 transcripts to DAVID Annotation Clustering analysis to derive their shared biological meaning. Interestingly, the MAPK signaling pathway and more specifically the ERK1/2 pathway geared toward cell survival/proliferation was significantly enriched. To conclude, we advocate incorporating pathway analysis into all microarray data analysis in order to improve the detection of similarities between independently derived datasets.
Collapse
Affiliation(s)
- K E M Cox-Limpens
- School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands; Department of Pediatrics, Maastricht University Medical Center (MUMC), postbus 5800, 6202 AZ Maastricht, The Netherlands.
| | - A W D Gavilanes
- Department of Pediatrics, Maastricht University Medical Center (MUMC), postbus 5800, 6202 AZ Maastricht, The Netherlands.
| | - L J I Zimmermann
- Department of Pediatrics, Maastricht University Medical Center (MUMC), postbus 5800, 6202 AZ Maastricht, The Netherlands.
| | - J S H Vles
- Department of Pediatric Neurology, Maastricht University Medical Center (MUMC), P.Debyelaan 25, 6229 HX Maastricht, The Netherlands.
| |
Collapse
|
43
|
Tsafnat G, Jasch D, Misra A, Choong MK, Lin FPY, Coiera E. Gene-disease association with literature based enrichment. J Biomed Inform 2014; 49:221-6. [PMID: 24681202 DOI: 10.1016/j.jbi.2014.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 02/09/2014] [Accepted: 03/02/2014] [Indexed: 10/25/2022]
Abstract
MOTIVATION Gene set enrichment analysis (GSEA) annotates gene microarray data with functional information from the biomedical literature to improve gene-disease association prediction. We hypothesize that supplementing GSEA with comprehensive gene function catalogs built automatically using information extracted from the scientific literature will significantly enhance GSEA prediction quality. METHODS Gold standard gene sets for breast cancer (BrCa) and colorectal cancer (CRC) were derived from the literature. Two gene function catalogs (CMeSH and CUMLS) were automatically generated. 1. By using Entrez Gene to associate all recorded human genes with PubMed article IDs. 2. Using the genes mentioned in each PubMed article and associating each with the article's MeSH terms (in CMeSH) and extracted UMLS concepts (in CUMLS). Microarray data from the Gene Expression Omnibus for BrCa and CRC was then annotated using CMeSH and CUMLS and for comparison, also with several pre-existing catalogs (C2, C4 and C5 from the Molecular Signatures Database). Ranking was done using, a standard GSEA implementation (GSEA-p). Gene function predictions for enriched array data were evaluated against the gold standard by measuring area under the receiver operating characteristic curve (AUC). RESULTS Comparison of ranking using the literature enrichment catalogs, the pre-existing catalogs as well as five randomly generated catalogs show the literature derived enrichment catalogs are more effective. The AUC for BrCa using the unenriched gene expression dataset was 0.43, increasing to 0.89 after gene set enrichment with CUMLS. The AUC for CRC using the unenriched gene expression dataset was 0.54, increasing to 0.9 after enrichment with CMeSH. C2 increased AUC (BrCa 0.76, CRC 0.71) but C4 and C5 performed poorly (between 0.35 and 0.5). The randomly generated catalogs also performed poorly, equivalent to random guessing. DISCUSSION Gene set enrichment significantly improved prediction of gene-disease association. Selection of enrichment catalog had a substantial effect on prediction accuracy. The literature based catalogs performed better than the MSigDB catalogs, possibly because they are more recent. Catalogs generated automatically from the literature can be kept up to date. CONCLUSION Prediction of gene-disease association is a fundamental task in biomedical research. GSEA provides a promising method when using literature-based enrichment catalogs. AVAILABILITY The literature based catalogs generated and used in this study are available from http://www2.chi.unsw.edu.au/literature-enrichment.
Collapse
Affiliation(s)
- Guy Tsafnat
- Centre for Health Informatics, University of New South Wales, Sydney, Australia.
| | - Dennis Jasch
- Centre for Health Informatics, University of New South Wales, Sydney, Australia
| | - Agam Misra
- Centre for Health Informatics, University of New South Wales, Sydney, Australia
| | - Miew Keen Choong
- Centre for Health Informatics, University of New South Wales, Sydney, Australia
| | - Frank P-Y Lin
- Centre for Health Informatics, University of New South Wales, Sydney, Australia
| | - Enrico Coiera
- Centre for Health Informatics, University of New South Wales, Sydney, Australia
| |
Collapse
|
44
|
O'Connor ML, Xiang D, Shigdar S, Macdonald J, Li Y, Wang T, Pu C, Wang Z, Qiao L, Duan W. Cancer stem cells: A contentious hypothesis now moving forward. Cancer Lett 2014; 344:180-187. [PMID: 24333726 DOI: 10.1016/j.canlet.2013.11.012] [Citation(s) in RCA: 185] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/11/2013] [Accepted: 11/19/2013] [Indexed: 02/07/2023]
Abstract
Cancer stem cells are a progressive concept to account for the cell biological nature of cancer. Despite the controversies regarding the cancer stem cell model, it has the potential to provide a foundation for new innovative treatment targeting the roots of cancer. The last two years have witnessed exceptional progress in cancer stem cell research, in particular on solid tumours, which holds promise for improved treatment outcomes. Here, we review recent advances in cancer stem cell research, discuss challenges in the field and explore future strategies and opportunities in cancer stem cell studies to overcome resistance to chemotherapy.
Collapse
Affiliation(s)
- Michael L O'Connor
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia
| | - Dongxi Xiang
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia
| | - Sarah Shigdar
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia
| | - Joanna Macdonald
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia
| | - Yong Li
- Cancer Care Centre, St. George Hospital, and St. George Clinical School, Faculty of Medicine, University of New South Wales, Kensington, NSW 2052, Australia
| | - Tao Wang
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia
| | - Chunwen Pu
- Dalian Sixth People's Hospital, Dalian 116033, China
| | - Zhidong Wang
- Dalian Sixth People's Hospital, Dalian 116033, China
| | - Liang Qiao
- Storr Liver Unit, Westmead Millennium Institute, The University of Sydney at the Westmead Hospital, Westmead, NSW 2145, Australia.
| | - Wei Duan
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia.
| |
Collapse
|
45
|
Soneson C, Fontes M. Incorporation of gene exchangeabilities improves the reproducibility of gene set rankings. Comput Stat Data Anal 2014. [DOI: 10.1016/j.csda.2012.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
46
|
Stapelberg M, Zobalova R, Nguyen MN, Walker T, Stantic M, Goodwin J, Pasdar EA, Thai T, Prokopova K, Yan B, Hall S, de Pennington N, Thomas SR, Grant G, Stursa J, Bajzikova M, Meedeniya ACB, Truksa J, Ralph SJ, Ansorge O, Dong LF, Neuzil J. Indoleamine-2,3-dioxygenase elevated in tumor-initiating cells is suppressed by mitocans. Free Radic Biol Med 2014; 67:41-50. [PMID: 24145120 DOI: 10.1016/j.freeradbiomed.2013.10.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 10/02/2013] [Accepted: 10/02/2013] [Indexed: 01/07/2023]
Abstract
Tumor-initiating cells (TICs) often survive therapy and give rise to second-line tumors. We tested the plausibility of sphere cultures as models of TICs. Microarray data and microRNA data analysis confirmed the validity of spheres as models of TICs for breast and prostate cancer as well as mesothelioma cell lines. Microarray data analysis revealed the Trp pathway as the only pathway upregulated significantly in all types of studied TICs, with increased levels of indoleamine-2,3-dioxygenase-1 (IDO1), the rate-limiting enzyme of Trp metabolism along the kynurenine pathway. All types of TICs also expressed higher levels of the Trp uptake system consisting of CD98 and LAT1 with functional consequences. IDO1 expression was regulated via both transcriptional and posttranscriptional mechanisms, depending on the cancer type. Serial transplantation of TICs in mice resulted in gradually increased IDO1. Mitocans, represented by α-tocopheryl succinate and mitochondrially targeted vitamin E succinate (MitoVES), suppressed IDO1 in TICs. MitoVES suppressed IDO1 in TICs with functional mitochondrial complex II, involving transcriptional and posttranscriptional mechanisms. IDO1 increase and its suppression by VE analogues were replicated in TICs from primary human glioblastomas. Our work indicates that IDO1 is increased in TICs and that mitocans suppress the protein.
Collapse
Affiliation(s)
- Michael Stapelberg
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia.
| | - Renata Zobalova
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia; Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic
| | - Maria Nga Nguyen
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Tom Walker
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia; Department of Neurosurgery, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Marina Stantic
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Jacob Goodwin
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Elham Alizadeh Pasdar
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Thuan Thai
- Centre for Vascular Research, School of Medical Sciences, University of New South Wales, Sydney, 2052 NSW, Australia
| | - Katerina Prokopova
- Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic; Faculty of Science, Charles University, 11000 Prague 1, Czech Republic
| | - Bing Yan
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Susan Hall
- School of Pharmacy, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | | | - Shane R Thomas
- Centre for Vascular Research, School of Medical Sciences, University of New South Wales, Sydney, 2052 NSW, Australia
| | - Gary Grant
- School of Pharmacy, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Jan Stursa
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia; Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague 160 00, Czech Republic
| | - Martina Bajzikova
- Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic
| | - Adrian C B Meedeniya
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Jaroslav Truksa
- Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic
| | - Stephen J Ralph
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Olaf Ansorge
- Department of Neurosurgery, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Lan-Feng Dong
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Jiri Neuzil
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia; Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic.
| |
Collapse
|
47
|
Qureshi AT, Chen C, Shah F, Thomas-Porch C, Gimble JM, Hayes DJ. Human Adipose-Derived Stromal/Stem Cell Isolation, Culture, and Osteogenic Differentiation. Methods Enzymol 2014; 538:67-88. [DOI: 10.1016/b978-0-12-800280-3.00005-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
48
|
Calautti E. Akt modes of stem cell regulation: more than meets the eye? Discoveries (Craiova) 2013; 1:e8. [PMID: 32309540 PMCID: PMC6941558 DOI: 10.15190/d.2013.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Akt signaling regulates many cellular functions that are essential for the proper balance between self-renewal and differentiation of tissue-specific and embryonic stem cells (SCs). However, the roles of Akt and its downstream signaling in SC regulation are rather complex, as Akt activation can either promote SC self-renewal or depletion in a context-dependent manner. In this review we have evidenced three "modes" of Akt-dependent SC regulation, which can be exemplified by three different SC types. In particular, we will discuss: 1) the integration of Akt signaling within the "core" SC signaling circuitry in the maintenance of SC self-renewal and pluripotency (embryonic SCs); 2) quantitative changes in Akt signaling in SC metabolic activity and exit from quiescence (hematopoietic SCs); 3) qualitative changes of Akt signaling in SC regulation: signaling compartment-talization and isoform-specific functions of Akt proteins in SC self-renewal and differentiation (limbal-corneal keratinocyte SCs). These diverse modes of action are not to be intended as mutually exclusive. Rather, it is likely that Akt proteins participate with multiple parallel mechanisms to regulation of the same SC type. We propose that under specific circumstances dictated by distinct developmental stages, differentiation programs or tissue culture conditions, one mode of Akt action prevails over the others in determining SC fates.
Collapse
Affiliation(s)
- Enzo Calautti
- University of Turin, Department of Molecular Biotechnology and Health Sciences, Turin, Italy
| |
Collapse
|
49
|
Osada M, Singh VJ, Wu K, Sant’Angelo DB, Pezzano M. Label retention identifies a multipotent mesenchymal stem cell-like population in the postnatal thymus. PLoS One 2013; 8:e83024. [PMID: 24340075 PMCID: PMC3858364 DOI: 10.1371/journal.pone.0083024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/29/2013] [Indexed: 02/06/2023] Open
Abstract
Thymic microenvironments are essential for the proper development and selection of T cells critical for a functional and self-tolerant adaptive immune response. While significant turnover occurs, it is unclear whether populations of adult stem cells contribute to the maintenance of postnatal thymic epithelial microenvironments. Here, the slow cycling characteristic of stem cells and their property of label-retention were used to identify a K5-expressing thymic stromal cell population capable of generating clonal cell lines that retain the capacity to differentiate into a number of mesenchymal lineages including adipocytes, chondrocytes and osteoblasts suggesting a mesenchymal stem cell-like phenotype. Using cell surface analysis both culture expanded LRCs and clonal thymic mesenchymal cell lines were found to express Sca1, PDGFRα, PDGFRβ,CD29, CD44, CD49F, and CD90 similar to MSCs. Sorted GFP-expressing stroma, that give rise to TMSC lines, contribute to thymic architecture when reaggregated with fetal stroma and transplanted under the kidney capsule of nude mice. Together these results show that the postnatal thymus contains a population of mesenchymal stem cells that can be maintained in culture and suggests they may contribute to the maintenance of functional thymic microenvironments.
Collapse
Affiliation(s)
- Masako Osada
- Department of Biology, The City College of New York, CUNY, New York, New York, United States of America
| | - Varan J. Singh
- Department of Biology, The City College of New York, CUNY, New York, New York, United States of America
| | - Kenmin Wu
- Department of Biology, The City College of New York, CUNY, New York, New York, United States of America
| | - Derek B. Sant’Angelo
- Child Health Institute of New Jersey, Department of Pediatrics, Rutgers, Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
| | - Mark Pezzano
- Department of Biology, The City College of New York, CUNY, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
50
|
Grafi G. Stress cycles in stem cells/iPSCs development: implications for tissue repair. Biogerontology 2013; 14:603-8. [PMID: 23852045 DOI: 10.1007/s10522-013-9445-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 07/07/2013] [Indexed: 12/12/2022]
Abstract
Stem cells have become a major topic, both publicly and scientifically, owing to their potential to cure diseases and repair damaged tissues. Particular attention has been given to the so-called "induced pluripotent stem cells" (iPSCs) in which somatic cells are induced by the expression of transcription factor encoding transgenes-a methodology first established by Takahashi and Yamanaka (Cell 126:663-676, 2006)-to acquire pluripotent state. This methodology has captured researchers' imagination as a potential procedure to obtain patient-specific therapies while also solving both the problem of transplant rejection and the ethical concerns often raised regarding the use of embryonic stem cells in regenerative medicine. The study of the biology of stem cells/iPSCs, in recent years, has uncovered some fundamental weaknesses that undermine their potential use in transplantation therapies.
Collapse
Affiliation(s)
- Gideon Grafi
- French Associates Institute for Agriculture and Biotechnology of Drylands, Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, 84990, Midreshet Ben-Gurion, Israel,
| |
Collapse
|