201
|
Corradetti B, Taraballi F, Powell S, Sung D, Minardi S, Ferrari M, Weiner BK, Tasciotti E. Osteoprogenitor cells from bone marrow and cortical bone: understanding how the environment affects their fate. Stem Cells Dev 2015; 24:1112-23. [PMID: 25517215 DOI: 10.1089/scd.2014.0351] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bone is a dynamic organ where skeletal progenitors and hematopoietic cells share and compete for space. Presumptive mesenchymal stem cells (MSC) have been identified and harvested from the bone marrow (BM-MSC) and cortical bone fragments (CBF-MSC). In this study, we demonstrate that despite the cells sharing a common ancestor, the differences in the structural properties of the resident tissues affect cell behavior and prime them to react differently to stimuli. Similarly to the bone marrow, the cortical portion of the bone contains a unique subset of cells that stains positively for the common MSC-associated markers. These cells display different multipotent differentiation capability, clonogenic expansion, and immunosuppressive potential. In particular, when compared with BM-MSC, CBF-MSC are bigger in size, show a lower proliferation rate at early passages, have a greater commitment toward the osteogenic lineage, constitutively produce nitric oxide as a mediator for bone remodeling, and more readily respond to proinflammatory cytokines. Our data suggest that the effect of the tissue's microenvironment makes the CBF-MSC a superior candidate in the development of new strategies for bone repair.
Collapse
Affiliation(s)
- Bruna Corradetti
- 1 Department of Nanomedicine, Houston Methodist Research Institute , Houston, Texas
| | | | | | | | | | | | | | | |
Collapse
|
202
|
Oxidative stress tolerance of early stage diabetic endothelial progenitor cell. Regen Ther 2015; 1:38-44. [PMID: 31245440 PMCID: PMC6581786 DOI: 10.1016/j.reth.2014.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 11/01/2014] [Accepted: 11/05/2014] [Indexed: 12/23/2022] Open
Abstract
Introduction One of the causes for poor vasculogenesis of diabetes mellitus (DM) is known to rise from the dysfunction of bone marrow-derived endothelial progenitor cells (BM EPCs). However, the origin of its cause is less understood. We aimed to investigate the effect of oxidative stress in early stage of diabetic BM-EPC and whether its vasculogenic dysfunction is caused by oxidative stress. Methods Bone marrow c-Kit+Sca-1+Lin− (BM-KSL) cells were sorted from control and streptozotocin-induced diabetic C57BL6J mice by flow cytometry. BM-KSLs were then assessed for vasculogenic potential (colony forming assay; EPC-CFA), accumulation of intracellular ROS (CM-H2DCFDA), carbonylated protein (ELISA), anti-oxidative enzymes expression (RT-qPCR) and catalase activity (Amplex Red). Results Compared to control, DM BM-KSL had significantly lower EPC-CFUs in both definitive EPC-CFU and total EPC-CFU (p < 0.05). Interestingly, the oxidative stress level of DM BM-KSL was comparable and was not significantly different to control followed by increased in anti-oxidative enzymes expression and catalase activity. Conclusions Primitive BM-EPCs showed vasculogenic dysfunction in early diabetes. However the oxidative stress is not denoted as the major initiating factor of its cause. Our results suggest that primitive BM-KSL cell has the ability to compensate oxidative stress levels in early diabetes by increasing the expression of anti-oxidative enzymes. Primitive BM-EPC showed EPC-CFU dysfunction in early diabetes. Primitive BM-EPC has the ability to withstand oxidative stress in early diabetes. Early diabetic BM-EPC increased anti-oxidative expression to compensate oxidative stress.
Collapse
|
203
|
Ozturk S, Karagoz H. Experimental stem cell therapies on burn wound: do source, dose, timing and method matter? Burns 2015; 41:1133-9. [PMID: 25716759 DOI: 10.1016/j.burns.2015.01.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 12/15/2014] [Accepted: 01/13/2015] [Indexed: 12/11/2022]
Abstract
Stem cell therapy has been introduced as a new and promising modality of wound covering in recent decade. It has been used for improvement of burn wound, post burn scar and saving stasis zone of burn with good results. However, there have been some differences between the various experimental burn wound trials in stem cell source, therapeutic dose, delivery method and timing of stem cell delivery. In our study, we aimed to review stem cell biology and investigate discrepancies in animal trials of use of stem cells in burn wound account for the variation in, stem cell source, therapeutic dose, delivery method and timing of stem cell delivery.
Collapse
Affiliation(s)
- Sinan Ozturk
- Gulhane Military Medical Academy, Haydarpasa Training Hospital, Plastic and Reconstructive Surgery Department, Turkey.
| | - Huseyin Karagoz
- Gulhane Military Medical Academy, Haydarpasa Training Hospital, Plastic and Reconstructive Surgery Department, Turkey
| |
Collapse
|
204
|
Abstract
The hemostatic system plays pleiotropic roles in cancer progression by shaping the tumor microenvironment and metastatic niches through thrombin-dependent fibrin deposition and platelet activation. Expanding experimental evidence implicates coagulation protease receptors expressed by tumor cells as additional players that directly influence tumor biology. Pro-angiogenic G protein-coupled signaling of TF through protease activated receptor 2 and regulation of tumor cell and vascular integrins through ligation by alternative spliced TF are established pathways driving tumor progression. Our recent work shows that the endothelial protein C receptor (EPCR), a stem cell marker in hematopoietic, neuronal and epithelial cells, is also crucial for breast cancer growth in the orthotopic microenvironment of the mammary gland. In aggressive triple-negative breast cancer cells, EPCR expression is a characteristic of cancer stem cell-like populations that have tumor initiating properties in vivo. Blocking antibodies to EPCR attenuate in vivo tumor growth and proliferation specifically of EPCR(+) cells on defined integrin matrices in vitro. We also showed that tumor-associated macrophages are a source for upstream coagulation proteases that can activate TF- and EPCR-dependent cellular responses, suggesting that tumor cells utilize the tumor microenvironment for tumor promoting coagulation protease signaling.
Collapse
|
205
|
Murtaza M, Jolly LA, Gecz J, Wood SA. La FAM fatale: USP9X in development and disease. Cell Mol Life Sci 2015; 72:2075-89. [PMID: 25672900 PMCID: PMC4427618 DOI: 10.1007/s00018-015-1851-0] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 02/02/2015] [Accepted: 02/04/2015] [Indexed: 11/12/2022]
Abstract
Deubiquitylating enzymes (DUBs), act downstream of ubiquitylation. As such, these post-post-translational modifiers function as the final arbitrators of a protein substrate’s ubiquitylation status, thus regulating its fate. In most instances, DUBs moderate the absolute level of a substrate, its locality or activity, rather than being an “all-or-none” phenomenon. Yet, disruption of this quantitative regulation can produce dramatic qualitative differences. The ubiquitin-specific protease 9X (USP9X/FAM) is a substrate-specific DUB, which displays an extraordinarily high level of sequence conservation from Drosophila to mammals. It is primarily the recent revelations of USP9X’s pivotal role in human cancers, both as oncogene or tumour suppressor, in developmental disorders including intellectual disability, epilepsy, autism and developmental delay that has led to a subsequent re-examination of its molecular and cellular functions. Results from experimental animal models have implicated USP9X in neurodegeneration, including Parkinson’s and Alzheimer’s disease, as well as autoimmune diseases. In this review, we describe the current and accumulated knowledge on the molecular, cellular and developmental aspects of USP9X function within the context of the biological consequences during normal development and disease.
Collapse
Affiliation(s)
- Mariyam Murtaza
- The Eskitis Institute for Drug Discovery, Griffith University, Brisbane, QLD, Australia
| | | | | | | |
Collapse
|
206
|
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
|
207
|
Yao R, Wong JY. The effects of mechanical stimulation on controlling and maintaining marrow stromal cell differentiation into vascular smooth muscle cells. J Biomech Eng 2015; 137:020907. [PMID: 25429403 DOI: 10.1115/1.4029255] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Indexed: 12/21/2022]
Abstract
For patients suffering from severe coronary heart disease (CHD), the development of a cell-based tissue engineered blood vessel (TEBV) has great potential to overcome current issues with synthetic graft materials. While marrow stromal cells (MSCs) are a promising source of vascular smooth muscle cells (VSMCs) for TEBV construction, they have been shown to differentiate into both the VSMC and osteoblast lineages under different rates of dynamic strain. Determining the permanence of strain-induced MSC differentiation into VSMCs is therefore a significant step toward successful TEBV development. In this study, initial experiments where a cyclic 10% strain was imposed on MSCs for 24 h at 0.1 Hz, 0.5 Hz, and 1 Hz determined that cells stretched at 1 Hz expressed significantly higher levels of VSMC-specific genetic and protein markers compared to samples stretched at 0.1 Hz. Conversely, samples stretched at 0.1 Hz expressed higher levels of osteoblast-specific genetic and protein markers compared to the samples stretched at 1 Hz. More importantly, sequential application of 24-48 h periods of 0.1 Hz and 1 Hz strain-induced genetic and protein marker expression levels similar to the VSMC profile seen with 1 Hz alone. This effect was observed regardless of whether the cells were first strained at 0.1 Hz followed by strain at 1 Hz, or vice versa. Our results suggest that the strain-induced VSMC phenotype is a more terminally differentiated state than the strain-induced osteoblast phenotype, and as result, VSMC obtained from strain-induced differentiation would have potential uses in TEBV construction.
Collapse
|
208
|
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease caused by aberrant proliferation and/or differentiation of myeloid progenitors. However, only ~65% of AML patients respond to induction chemotherapy and the overall survival rate for AML remains low (~24% for 5-year survival). The conventional view suggests that ATP-binding cassette (ABC) transporters contribute to treatment failure due to their drug-effluxing capabilities. This might be overly simplistic. Some ABC transporters export endogenous substrates that have defined roles in normal hematopoietic progenitors. It is conceivable that these substances also provide an advantage to leukemic progenitors. This review will highlight how certain endogenous substrates impact normal hematopoietic cells and suggest that ABC transporters facilitate export of these substances to affect both normal hematopoietic and leukemic progenitors. For example, the ability to export certain endogenous ligands may facilitate leukemogenesis by modifying leukemic progenitor cell proliferation or survival. If so, the addition of ABC transporter inhibitors to traditional chemotherapy might improve therapeutic efficacy by not just increasing intracellular drug accumulation but also blocking the beneficial effects ABC transporter ligands have on cell survival.
Collapse
|
209
|
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
|
210
|
Park MS, Kausar R, Kim MW, Cho SY, Lee YS, Lee MA. Tcf7l1-mediated transcriptional regulation of Krüppel-like factor 4 gene. Anim Cells Syst (Seoul) 2015. [DOI: 10.1080/19768354.2014.991351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
211
|
Gro/TLE enables embryonic stem cell differentiation by repressing pluripotent gene expression. Dev Biol 2015; 397:56-66. [DOI: 10.1016/j.ydbio.2014.10.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 09/21/2014] [Accepted: 10/14/2014] [Indexed: 01/03/2023]
|
212
|
Chen F, Zhang X, Bi D, Xia L, Lin Y, Zhang W, Liu W, Cao Y. RETRACTED ARTICLE: Screening research on membrane protein of dermal stem/progenitor cells with different differentiation potential. Cytotechnology 2015; 67:175. [PMID: 23475157 PMCID: PMC4294848 DOI: 10.1007/s10616-012-9475-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Accepted: 06/08/2012] [Indexed: 10/27/2022] Open
Affiliation(s)
- Fuguo Chen
- />Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, No.639 Zhi Zaoju Road, Shanghai, 200011 People’s Republic of China
| | - Xiaoping Zhang
- />Department of Nuclear Medicine, Shanghai 10th People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 People’s Republic of China
| | - Dan Bi
- />Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, No.639 Zhi Zaoju Road, Shanghai, 200011 People’s Republic of China
| | - Linlin Xia
- />Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, No.639 Zhi Zaoju Road, Shanghai, 200011 People’s Republic of China
| | - Yang Lin
- />Department of Nuclear Medicine, Shanghai 10th People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 People’s Republic of China
| | - Wenjie Zhang
- />Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, No.639 Zhi Zaoju Road, Shanghai, 200011 People’s Republic of China
| | - Wei Liu
- />Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, No.639 Zhi Zaoju Road, Shanghai, 200011 People’s Republic of China
| | - Yilin Cao
- />Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, No.639 Zhi Zaoju Road, Shanghai, 200011 People’s Republic of China
| |
Collapse
|
213
|
Stem Cells in Regenerative Therapy. Bioengineering (Basel) 2015. [DOI: 10.1007/978-3-319-10798-1_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
214
|
Burroughs AM, Aravind L. Analysis of two domains with novel RNA-processing activities throws light on the complex evolution of ribosomal RNA biogenesis. Front Genet 2014; 5:424. [PMID: 25566315 PMCID: PMC4275035 DOI: 10.3389/fgene.2014.00424] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 11/19/2014] [Indexed: 11/13/2022] Open
Abstract
Ribosomal biogenesis has been extensively investigated, especially to identify the elusive nucleases and cofactors involved in the complex rRNA processing events in eukaryotes. Large-scale screens in yeast identified two biochemically uncharacterized proteins, TSR3 and TSR4, as being key players required for rRNA maturation. Using multiple computational approaches we identify the conserved domains comprising these proteins and establish sequence and structural features providing novel insights regarding their roles. TSR3 is unified with the DTW domain into a novel superfamily of predicted enzymatic domains, with the balance of the available evidence pointing toward an RNase role with the archaeo-eukaryotic TSR3 proteins processing rRNA and the bacterial versions potentially processing tRNA. TSR4, its other eukaryotic homologs PDCD2/rp-8, PDCD2L, Zfrp8, and trus, the predominantly bacterial DUF1963 proteins, and other uncharacterized proteins are unified into a new domain superfamily, which arose from an ancient duplication event of a strand-swapped, dimer-forming all-beta unit. We identify conserved features mediating protein-protein interactions (PPIs) and propose a potential chaperone-like function. While contextual evidence supports a conserved role in ribosome biogenesis for the eukaryotic TSR4-related proteins, there is no evidence for such a role for the bacterial versions. Whereas TSR3-related proteins can be traced to the last universal common ancestor (LUCA) with a well-supported archaeo-eukaryotic branch, TSR4-related proteins of eukaryotes are derived from within the bacterial radiation of this superfamily, with archaea entirely lacking them. This provides evidence for “systems admixture,” which followed the early endosymbiotic event, playing a key role in the emergence of the uniquely eukaryotic ribosome biogenesis process.
Collapse
Affiliation(s)
- A Maxwell Burroughs
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health Bethesda, MD, USA
| | - L Aravind
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health Bethesda, MD, USA
| |
Collapse
|
215
|
Peng Y, Xuan M, Zou J, Liu H, Zhuo Z, Wan Y, Cheng B. Freeze-dried rat bone marrow mesenchymal stem cell paracrine factors: a simplified novel material for skin wound therapy. Tissue Eng Part A 2014; 21:1036-46. [PMID: 25343727 DOI: 10.1089/ten.tea.2014.0102] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The mesenchymal stem cell (MSC) supernatant is well known as a rich source of autologous cytokines and universally used for tissue regeneration in current clinical medicine. However, the limitation of conditioned medium used in open-wound repair compels the need to find a more sophisticated way to take advantage of the trophic factors of MSCs. We have now fabricated a three-dimensional membrane from freeze-dried bone marrow mesenchymal stem cells-conditioned medium (FBMSC-CM) using a simple freeze-dried protocol. Scanning electron microscopy images showed the microstructure of the FBMSC-CM membrane (FBMSC-CMM) resembling a mesh containing growth factors. ELISA was used to test the paracrine factors retained in the FBMSC-CMM, and the results indicated that FBMSC-CMM withheld over 80% of the paracrine factors. Live/dead assays were adopted to test the toxicity of the FBMSC-CMM on cultured rat dermal fibroblasts, and the results confirmed its biological safety with low toxicity. Moreover, the FBMSC-CMM could significantly accelerate wound healing and enhance the neovascularization as well as epithelialization through strengthening the trophic factors in the wound bed as determined by immunohistochemical staining. Thus, the ability to maintain paracrine factors and enhance the effectiveness of these growth factors in the wound as well as the simple procedure and economical materials required for production qualifies the FBMSC-CMM to be a candidate biomaterial for open-wound regeneration.
Collapse
Affiliation(s)
- Yan Peng
- 1 Department of Plastic Surgery, Guangzhou General Hospital of Guangzhou Command , The Key Laboratory of Trauma Treatment & Tissue Repair of Tropical Area, PLA, Guangzhou, P.R. China
| | | | | | | | | | | | | |
Collapse
|
216
|
Veeriah V, Saran U, Swaminathan A, Balaguru UM, Thangaraj P, Nagarajan S, Rajendran VK, Chatterjee S. Cadmium-induced embryopathy: nitric oxide rescues teratogenic effects of cadmium. Toxicol Sci 2014; 144:90-104. [PMID: 25490952 DOI: 10.1093/toxsci/kfu258] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although Cadmium (Cd) is a well-known heavy metal pollutant and teratogen, the mechanism behind Cd-mediated teratogenicity remains unknown. Previously, we have reported of the protective role of Nitric oxide (NO), a key signaling molecule in the embryonic developmental process, against Thalidomide-induced teratogenicity. The objective of this study was to obtain a mechanistic in-sight of the antiteratogenic potential of NO against Cd-mediated teratogenicity. To achieve this goal, we first studied the effect of Cd on the vasculature of developing embryos and then we investigated whether Cd mediated its effects by interfering with the redox regulation of NO signaling in the early development milieu. We used a chick embryonic model to determine the time and dose-dependent effects of Cd and NO recovery against Cd assault. The effects of Cd and NO recovery were assessed using various angiogenic assays. Redox and NO levels were also measured. Results demonstrated that exposure to Cd at early stage of development caused multiple birth defects in the chick embryos. Exposure to Cd suppressed endogenous NO levels and cGMP signaling, inhibiting angioblast activation and subsequently impairing yolk sac vascular development. Furthermore, Cd-induced superoxide and lipid peroxidation mediated activation of proapoptotic markers p21 and p53 in the developing embryo. Cd also caused the down-regulation of FOXO1, and up-regulation of FOXO3a and Caspase 3-mediated apoptosis. Addition of exogenous NO through a NO donor was able to blunt Cd-mediated effects and restore normal vascular and embryonic development. In conclusion, Cd-mediated teratogenicity occurs as a result of impaired NO-cGMP signaling, increased oxidative stress, and the activation of apoptotic pathways. Subsequent addition of exogenous NO through NO donor negated Cd-mediated effects and protected the developing embryo.
Collapse
Affiliation(s)
- Vimal Veeriah
- *Vascular Biology Laboratory, AU-KBC Research Centre, Anna University, Chennai 600044, India and Department of Biotechnology, Anna University, Chennai 600044, India
| | - Uttara Saran
- *Vascular Biology Laboratory, AU-KBC Research Centre, Anna University, Chennai 600044, India and Department of Biotechnology, Anna University, Chennai 600044, India
| | - Akila Swaminathan
- *Vascular Biology Laboratory, AU-KBC Research Centre, Anna University, Chennai 600044, India and Department of Biotechnology, Anna University, Chennai 600044, India
| | - Uma Maheswari Balaguru
- *Vascular Biology Laboratory, AU-KBC Research Centre, Anna University, Chennai 600044, India and Department of Biotechnology, Anna University, Chennai 600044, India
| | - Pradeep Thangaraj
- *Vascular Biology Laboratory, AU-KBC Research Centre, Anna University, Chennai 600044, India and Department of Biotechnology, Anna University, Chennai 600044, India
| | - Shunmugam Nagarajan
- *Vascular Biology Laboratory, AU-KBC Research Centre, Anna University, Chennai 600044, India and Department of Biotechnology, Anna University, Chennai 600044, India
| | - Vinoth Kumar Rajendran
- *Vascular Biology Laboratory, AU-KBC Research Centre, Anna University, Chennai 600044, India and Department of Biotechnology, Anna University, Chennai 600044, India
| | - Suvro Chatterjee
- *Vascular Biology Laboratory, AU-KBC Research Centre, Anna University, Chennai 600044, India and Department of Biotechnology, Anna University, Chennai 600044, India *Vascular Biology Laboratory, AU-KBC Research Centre, Anna University, Chennai 600044, India and Department of Biotechnology, Anna University, Chennai 600044, India
| |
Collapse
|
217
|
Guzman-Ayala M, Sachs M, Koh FM, Onodera C, Bulut-Karslioglu A, Lin CJ, Wong P, Nitta R, Song JS, Ramalho-Santos M. Chd1 is essential for the high transcriptional output and rapid growth of the mouse epiblast. Development 2014; 142:118-27. [PMID: 25480920 DOI: 10.1242/dev.114843] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The pluripotent mammalian epiblast undergoes unusually fast cell proliferation. This rapid growth is expected to generate a high transcriptional demand, but the underlying mechanisms remain unknown. We show here that the chromatin remodeler Chd1 is required for transcriptional output and development of the mouse epiblast. Chd1(-/-) embryos exhibit proliferation defects and increased apoptosis, are smaller than controls by E5.5 and fail to grow, to become patterned or to gastrulate. Removal of p53 allows progression of Chd1(-/-) mutants only to E7.0-8.0, highlighting the crucial requirement for Chd1 during early post-implantation development. Chd1(-/-) embryonic stem cells (ESCs) have a self-renewal defect and a genome-wide reduction in transcriptional output at both known mRNAs and intergenic transcripts. These transcriptional defects were only uncovered when cell number-normalized approaches were used, and correlate with a lower engagement of RNAP II with transcribed genes in Chd1(-/-) ESCs. We further show that Chd1 directly binds to ribosomal DNA, and that both Chd1(-/-) epiblast cells in vivo and ESCs in vitro express significantly lower levels of ribosomal RNA. In agreement with these findings, mutant cells in vivo and in vitro exhibit smaller and more elongated nucleoli. Thus, the RNA output by both Pol I and II is reduced in Chd1(-/-) cells. Our data indicate that Chd1 promotes a globally elevated transcriptional output required to sustain the distinctly rapid growth of the mouse epiblast.
Collapse
Affiliation(s)
- Marcela Guzman-Ayala
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA
| | - Michael Sachs
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA
| | - Fong Ming Koh
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA
| | - Courtney Onodera
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA Institute for Human Genetics, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA Department of Epidemiology and Biostatistics, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA
| | - Aydan Bulut-Karslioglu
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA
| | - Chih-Jen Lin
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA
| | - Priscilla Wong
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA
| | - Rachel Nitta
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA
| | - Jun S Song
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA Institute for Human Genetics, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA Department of Epidemiology and Biostatistics, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA
| | - Miguel Ramalho-Santos
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA Center for Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, 35 Medical Center Way, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
218
|
Zhang S, Liu P, Chen L, Wang Y, Wang Z, Zhang B. The effects of spheroid formation of adipose-derived stem cells in a microgravity bioreactor on stemness properties and therapeutic potential. Biomaterials 2014; 41:15-25. [PMID: 25522961 DOI: 10.1016/j.biomaterials.2014.11.019] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/30/2014] [Accepted: 11/07/2014] [Indexed: 02/07/2023]
Abstract
Adipose-derived stem cells (ADSCs) represent a valuable source of stem cells for regenerative medicine, but the loss of their stemness during in vitro expansion remains a major roadblock. We employed a microgravity bioreactor (MB) to develop a method for biomaterial-free-mediated spheroid formation to maintain the stemness properties of ADSCs. ADSCs spontaneously formed three-dimensional spheroids in the MB. Compared with monolayer culture, the expression levels of E-cadherin and pluripotent markers were significantly upregulated in ADSC spheroids. Spheroid-derived ADSCs exhibited increased proliferative ability and colony-forming efficiency. By culturing the spheroid-derived ADSCs in an appropriate induction medium, we found that the multipotency differentiation capacities of ADSCs were significantly improved by spheroid culture in the MB. Furthermore, when ADSCs were administered to mice with carbon tetrachloride-induced acute liver failure, spheroid-derived ADSCs showed more effective potentials to rescue liver failure than ADSCs derived from constant monolayer culture. Our results suggest that spheroid formation of ADSCs in an MB enhances their stemness properties and increases their therapeutic potential. Therefore, spheroid culture in an MB can be an efficient method to maintain stemness properties, without the involvement of any biomaterials for clinical applications of in vitro cultured ADSCs.
Collapse
Affiliation(s)
- Shichang Zhang
- Department 4, Institute of Surgery Research, Daping Hospital, Third Military Medical University, State Key Lab of Trauma, Burns and Combined Injury, Chongqing 400042, China
| | - Ping Liu
- Department 4, Institute of Surgery Research, Daping Hospital, Third Military Medical University, State Key Lab of Trauma, Burns and Combined Injury, Chongqing 400042, China
| | - Li Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210017, China
| | - Yingjie Wang
- Institute of Infectious Diseases, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Zhengguo Wang
- Department 4, Institute of Surgery Research, Daping Hospital, Third Military Medical University, State Key Lab of Trauma, Burns and Combined Injury, Chongqing 400042, China
| | - Bo Zhang
- Department 4, Institute of Surgery Research, Daping Hospital, Third Military Medical University, State Key Lab of Trauma, Burns and Combined Injury, Chongqing 400042, China.
| |
Collapse
|
219
|
Khalaj M, Tavakkoli M, Stranahan AW, Park CY. Pathogenic microRNA's in myeloid malignancies. Front Genet 2014; 5:361. [PMID: 25477897 PMCID: PMC4237136 DOI: 10.3389/fgene.2014.00361] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 09/27/2014] [Indexed: 12/21/2022] Open
Abstract
Recent studies have significantly improved our understanding of the role microRNAs (miRNAs) play in regulating normal hematopoiesis. miRNAs are critical for maintaining hematopoietic stem cell function and the development of mature progeny. Thus, perhaps it is not surprising that miRNAs serve as oncogenes and tumor suppressors in hematologic malignancies arising from hematopoietic stem and progenitor cells, such as the myeloid disorders. A number of studies have extensively documented the widespread dysregulation of miRNA expression in human acute myeloid leukemia (AML), inspiring numerous explorations of the functional role of miRNAs in myeloid leukemogenesis. While these investigations have confirmed that a large number of miRNAs exhibit altered expression in AML, only a small fraction has been confirmed as functional mediators of AML development or maintenance. Herein, we summarize the miRNAs for which strong experimental evidence supports their functional roles in AML pathogenesis. We also discuss the implications of these studies on the development of miRNA-directed therapies in AML.
Collapse
Affiliation(s)
- Mona Khalaj
- Weill Graduate School of Medical Sciences, Cornell University NY, USA ; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center NY, USA
| | - Montreh Tavakkoli
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center NY, USA
| | - Alec W Stranahan
- Weill Graduate School of Medical Sciences, Cornell University NY, USA ; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center NY, USA
| | - Christopher Y Park
- Weill Graduate School of Medical Sciences, Cornell University NY, USA ; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center NY, USA ; Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center NY, USA
| |
Collapse
|
220
|
Fong YW, Ho JJ, Inouye C, Tjian R. The dyskerin ribonucleoprotein complex as an OCT4/SOX2 coactivator in embryonic stem cells. eLife 2014; 3. [PMID: 25407680 PMCID: PMC4270071 DOI: 10.7554/elife.03573] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 11/19/2014] [Indexed: 01/06/2023] Open
Abstract
Acquisition of pluripotency is driven largely at the transcriptional level by activators OCT4, SOX2, and NANOG that must in turn cooperate with diverse coactivators to execute stem cell-specific gene expression programs. Using a biochemically defined in vitro transcription system that mediates OCT4/SOX2 and coactivator-dependent transcription of the Nanog gene, we report the purification and identification of the dyskerin (DKC1) ribonucleoprotein complex as an OCT4/SOX2 coactivator whose activity appears to be modulated by a subset of associated small nucleolar RNAs (snoRNAs). The DKC1 complex occupies enhancers and regulates the expression of key pluripotency genes critical for self-renewal in embryonic stem (ES) cells. Depletion of DKC1 in fibroblasts significantly decreased the efficiency of induced pluripotent stem (iPS) cell generation. This study thus reveals an unanticipated transcriptional role of the DKC1 complex in stem cell maintenance and somatic cell reprogramming. DOI:http://dx.doi.org/10.7554/eLife.03573.001 The stem cells found in an embryo are able to develop into any of the cell types found in the body of the animal: an ability called pluripotency. When a cell becomes a specialized cell type, such as a nerve cell or a muscle cell, it loses this ability. However, mature cells can be reprogrammed back to a pluripotent state by artificially introducing certain proteins (known as ‘reprogramming factors’) into the mature cells. A core group of reprogramming factors are known to activate networks of genes that are normally only expressed in stem cells, and by doing so trigger and maintain a pluripotent state. Other proteins help these core factors to regulate these networks of genes. In 2011, researchers discovered that a protein complex called XPC—which is normally involved in DNA repair—also helps two core reprogramming factors to activate an important gene related to pluripotency. Now, Fong et al., including several of the researchers involved in the 2011 work, have identified another unexpected partner for the same two core reprogramming factors. The protein complex, called DKC1, has a number of known functions related to the processing of RNA molecules. This complex has also been linked to a fatal, rare human disorder called dyskeratosis congenita—a condition that affects many parts of the body, including the skin and bone marrow. Fong et al. found that when embryonic stems cells from mice are depleted of the DKC1 complex, the activation of important pluripotency-related genes by two of the core reprogramming factors is markedly reduced. The XPC and DKC1 protein complexes were found to interact in pluripotent cells, and together they can activate a pluripotency-related gene to a greater extent than either can individually. Fong et al. propose that DKC1 binds to XPC, which in turn binds to two of the core reprogramming factors. The DKC1 complex also binds to RNA molecules, and Fong et al. found that when the DKC1 complex binds to certain RNAs it is more able to help reprogramming factors activate pluripotency-related genes. On the other hand, other RNA molecules seem to inhibit the complex's ability to activate these genes. Mutations identified in people with dyskeratosis congenita can prevent the DKC1 complex from binding to a subset of human RNA molecules. Moreover, the activity of stem cells is impaired in people with this developmental condition. As such, one of the next challenges will be to investigate if these mutations and RNA binding could be linked to problems with the activation of genes related to pluripotency in stem cells. DOI:http://dx.doi.org/10.7554/eLife.03573.002
Collapse
Affiliation(s)
- Yick W Fong
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Jaclyn J Ho
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Carla Inouye
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Robert Tjian
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
221
|
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
|
222
|
Ferguson GB, Martinez-Agosto JA. Yorkie and Scalloped signaling regulates Notch-dependent lineage specification during Drosophila hematopoiesis. Curr Biol 2014; 24:2665-72. [PMID: 25454586 DOI: 10.1016/j.cub.2014.09.081] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 07/25/2014] [Accepted: 09/01/2014] [Indexed: 01/02/2023]
Abstract
Cellular microenvironments established by the spatial and temporal expression of specific signaling molecules are critical for both the maintenance and lineage-specific differentiation of progenitor cells. In Drosophila, a population of hematopoietic progenitors, or prohemocytes, within the larval lymph gland gives rise to three mature cell types: plasmatocytes, lamellocytes, and crystal cells. Removal of the secreted signaling molecules Hedgehog and PVF1 from the posterior signaling center (PSC), which acts as a niche, leads to a loss of progenitors and complete differentiation of the lymph gland. Here, we characterize a novel population of signaling cells within the lymph gland, distinct from the PSC, that are required for lineage-specific differentiation of crystal cells. We provide evidence that Yorkie and Scalloped, the Drosophila homologs of YAP and TEAD, are required in lineage-specifying cells to regulate expression of Serrate, the Notch ligand responsible for the initiation of the crystal cell differentiation program. Genetic manipulation of yorkie and scalloped in the lymph gland specifically alters Serrate expression and crystal cell differentiation. Furthermore, Serrate expression in lineage-specifying cells is eliminated in the lymph gland upon the immune response induced by wasp parasitization to ensure the proper differentiation of lamellocytes at the expense of crystal cells. These findings expand the roles for Yorkie/Scalloped beyond growth to encompass specific cell-fate determination in the context of blood development. Similar regulatory functions may extend to their homologs in vertebrate progenitor cell niches that are required for specifying cell fate.
Collapse
Affiliation(s)
- Gabriel B Ferguson
- Molecular Biology Interdepartmental Ph.D. Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Julian A Martinez-Agosto
- Molecular Biology Interdepartmental Ph.D. Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, Jonsson Comprehensive Cancer Center, UCLA Broad Stem Cell Center, and Mattel Children's Hospital UCLA, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
223
|
The Membrane Associated RING-CH Proteins: A Family of E3 Ligases with Diverse Roles through the Cell. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:637295. [PMID: 27419207 PMCID: PMC4897099 DOI: 10.1155/2014/637295] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 08/22/2014] [Indexed: 01/03/2023]
Abstract
Since the discovery that conjugation of ubiquitin to proteins can drive proteolytic degradation, ubiquitination has been shown to perform a diverse range of functions in the cell. It plays an important role in endocytosis, signal transduction, trafficking of vesicles inside the cell, and even DNA repair. The process of ubiquitination-mediated control has turned out to be remarkably complex, involving a diverse array of proteins and many levels of control. This review focuses on a family of structurally related E3 ligases termed the membrane-associated RING-CH (MARCH) ubiquitin ligases, which were originally discovered as structural homologs to the virals E3s, K3, and K5 from Kaposi's sarcoma-associated herpesvirus (KSHV). These proteins contain a catalytic RING-CH finger and are typically membrane-bound, with some having up to 14 putative transmembrane domains. Despite several lines of evidence showing that the MARCH proteins play a complex and essential role in several cellular processes, this family remains understudied.
Collapse
|
224
|
Zhang J, Wei W, Jin HC, Ying RC, Zhu AK, Zhang FJ. Programmed cell death 2 protein induces gastric cancer cell growth arrest at the early S phase of the cell cycle and apoptosis in a p53-dependent manner. Oncol Rep 2014; 33:103-10. [PMID: 25334010 DOI: 10.3892/or.2014.3551] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 08/02/2014] [Indexed: 11/06/2022] Open
Abstract
Programmed cell death 2 (PDCD2) is a highly conserved nuclear protein, and aberrant PDCD2 expression alters cell apoptosis. The present study aimed to investigate PDCD2 expression in gastric cancer. Tissue specimens from 34 gastric cancer patients were collected for analysis of PDCD2 expression using immunohistochemistry, western blotting and qRT-PCR. Gastric cancer cell lines (a p53-mutated MKN28 line and a wild-type p53 MKN45 line) were used to assess the effects of PDCD2 overexpression. p53-/- nude mice were used to investigate the effect of PDCD2 on ultraviolet B (UVB)-induced skin carcinogenesis. The data showed that PDCD2 expression was reduced in gastric cancer tissue specimens, and loss of PDCD2 expression was associated with the poor survival of patients. PDCD2 expression induced gastric cancer cell growth arrest at the early S phase of the cell cycle and apoptosis. The antitumor effects of PDCD2 expression were dependent on p53 expression in gastric cancer cells. Moreover, PDCD2 expression inhibited activity of the ATM/Chk1/2/p53 signaling pathway. In addition, PDCD2 expression suppressed UVB-induced skin carcinogenesis in p53+/+ nude mice, but not in p53-/- mice. The data from the present study demonstrated that loss of PDCD2 expression could contribute to gastric cancer development and progression and that PDCD2-induced gastric cancer cell growth arrest at the early S phase of the cell cycle and apoptosis are p53-dependent.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - Wei Wei
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - Hui-Cheng Jin
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - Rong-Chao Ying
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - A-Kao Zhu
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| | - Fang-Jie Zhang
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital, School of Clinical Medicine, Nanjing Medical University, Hangzhou 310006, P.R. China
| |
Collapse
|
225
|
Lee DK, Yi T, Park KE, Lee HJ, Cho YK, Lee SJ, Lee J, Park JH, Lee MY, Song SU, Kwon SW. Non-invasive characterization of the adipogenic differentiation of human bone marrow-derived mesenchymal stromal cells by HS-SPME/GC-MS. Sci Rep 2014; 4:6550. [PMID: 25298091 PMCID: PMC4190506 DOI: 10.1038/srep06550] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 08/11/2014] [Indexed: 01/10/2023] Open
Abstract
A non-invasive method to characterize human mesenchymal stromal cells during adipogenic differentiation was developed for the first time. Seven fatty acid methyl esters (FAMEs), including methyl laurate, methyl myristate, methyl palmitate, methyl linoleate, methyl oleate, methyl elaidate and methyl stearate, were used for characterizing adipogenic differentiation using headspace solid-phase microextraction (HS-SPME) which is a very simple and non-invasive method for the extraction of volatile compounds. Glassware was used for culturing mesenchymal stromal cells rather than the common plasticware to minimize contamination by volatile impurities. The optimal SPME fiber was selected by comparing diverse fibers containing two pure liquid polymers (PDMS and PA) and two porous solids (PDMS/DVB and CAR/PDMS). Using optimized procedures, we discovered that seven FAMEs were only detected in adipogenic differentiated mesenchymal stromal cells and not in the mesenchymal stromal cells before differentiation. These data could support the quality control of clinical mesenchymal stromal cell culture in the pharmaceutical industry in addition to the development of many clinical applications using mesenchymal stromal cells.
Collapse
Affiliation(s)
- Dong-Kyu Lee
- 1] College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea [2]
| | - TacGhee Yi
- 1] Translational Research Center and Inha Research Institute for Medical Sciences, Inha University School of Medicine, Incheon 400-712, Republic of Korea [2]
| | - Kyung-Eun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Hyun-Joo Lee
- Drug Development Program, Department of Medicine, Inha University School of Medicine, Incheon 400-712, Republic of Korea
| | - Yun-Kyoung Cho
- Translational Research Center and Inha Research Institute for Medical Sciences, Inha University School of Medicine, Incheon 400-712, Republic of Korea
| | - Seul Ji Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Jeongmi Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Jeong Hill Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Mi-Young Lee
- Occupational Safety and Health Research Institute, Korea Occupational Safety and Health Agency Incheon 403-711, Republic of Korea
| | - Sun U Song
- Translational Research Center and Inha Research Institute for Medical Sciences, Inha University School of Medicine, Incheon 400-712, Republic of Korea
| | - Sung Won Kwon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| |
Collapse
|
226
|
Zhang S, Tian D, Tran NH, Choi KP, Zhang L. Profiling the transcription factor regulatory networks of human cell types. Nucleic Acids Res 2014; 42:12380-7. [PMID: 25300490 PMCID: PMC4227771 DOI: 10.1093/nar/gku923] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Neph et al. (2012) (Circuitry and dynamics of human transcription factor regulatory networks. Cell, 150: 1274-1286) reported the transcription factor (TF) regulatory networks of 41 human cell types using the DNaseI footprinting technique. This provides a valuable resource for uncovering regulation principles in different human cells. In this paper, the architectures of the 41 regulatory networks and the distributions of housekeeping and specific regulatory interactions are investigated. The TF regulatory networks of different human cell types demonstrate similar global three-layer (top, core and bottom) hierarchical architectures, which are greatly different from the yeast TF regulatory network. However, they have distinguishable local organizations, as suggested by the fact that wiring patterns of only a few TFs are enough to distinguish cell identities. The TF regulatory network of human embryonic stem cells (hESCs) is dense and enriched with interactions that are unseen in the networks of other cell types. The examination of specific regulatory interactions suggests that specific interactions play important roles in hESCs.
Collapse
Affiliation(s)
- Shihua Zhang
- National Center for Mathematics and Interdisciplinary Sciences, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
| | - Dechao Tian
- Department of Statistics and Applied Probability, National University of Singapore, Singapore 117546, Singapore
| | - Ngoc Hieu Tran
- Division of Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| | - Kwok Pui Choi
- Department of Statistics and Applied Probability, National University of Singapore, Singapore 117546, Singapore Department of Mathematics, National University of Singapore, Singapore 119076, Singapore
| | - Louxin Zhang
- Department of Mathematics, National University of Singapore, Singapore 119076, Singapore National University of Singapore Graduate School for Integrative Sciences and Engineering, Singapore 117456, Singapore
| |
Collapse
|
227
|
Knockdown of ZFX suppresses renal carcinoma cell growth and induces apoptosis. Cancer Genet 2014; 207:461-6. [DOI: 10.1016/j.cancergen.2014.08.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 08/28/2014] [Accepted: 08/30/2014] [Indexed: 02/02/2023]
|
228
|
Developmental pathways hijacked by osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 804:93-118. [PMID: 24924170 DOI: 10.1007/978-3-319-04843-7_5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cancer of any type often can be described by an arrest, alteration or disruption in the normal development of a tissue or organ, and understanding of the normal counterpart's development can aid in understanding the malignant state. This is certainly true for osteosarcoma and the normal developmental pathways that guide osteoblast development that are changed in the genesis of osteogenic sarcoma. A carefully regulated crescendo-decrescendo expression of RUNX2 accompanies the transition from mesenchymal stem cell to immature osteoblast to mature osteoblast. This pivotal role is controlled by several pathways, including bone morphogenic protein (BMP), Wnt/β-catenin, fibroblast growth factor (FGF), and protein kinase C (PKC). The HIPPO pathway and its downstream target YAP help to regulate proliferation of immature osteoblasts and their maturation into non-proliferating mature osteoblasts. This pathway also helps regulate expression of the mature osteoblast protein osteocalcin. YAP also regulates expression of MT1-MMP, a membrane-bound matrix metalloprotease responsible for remodeling the extracellular matrix surrounding the osteoblasts. YAP, in turn, can be regulated by the ERBB family protein Her-4. Osteosarcoma may be thought of as a cell held at the immature osteoblast stage, retaining some of the characteristics of that developmental stage. Disruptions of several of these pathways have been described in osteosarcoma, including BMP, Wnt/b-catenin, RUNX2, HIPPO/YAP, and Her-4. Further, PKC can be activated by several receptor tyrosine kinases implicated in osteosarcoma, including the ERBB family (EGFR, Her-2 and Her-4 in osteosarcoma), IGF1R, FGF, and others. Understanding these functions may aid in the understanding the mechanisms underpinning clinical observations in osteosarcoma, including both the lytic and blastic phenotypes of tumors, the invasiveness of the disease, and the tendency for treated tumors to ossify rather than shrink. Through a better understanding of the relationship between normal osteoblast development and osteosarcoma, we may gain insights into novel therapeutic avenues and improved outcomes.
Collapse
|
229
|
Mavila N, James D, Shivakumar P, Nguyen MV, Utley S, Mak K, Wu A, Zhou S, Wang L, Vendyres C, Groff M, Asahina K, Wang KS. Expansion of prominin-1-expressing cells in association with fibrosis of biliary atresia. Hepatology 2014; 60:941-953. [PMID: 24798639 PMCID: PMC4146699 DOI: 10.1002/hep.27203] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 04/24/2014] [Accepted: 04/29/2014] [Indexed: 02/06/2023]
Abstract
UNLABELLED Biliary atresia (BA), the most common cause of end-stage liver disease and the leading indication for pediatric liver transplantation, is associated with intrahepatic ductular reactions within regions of rapidly expanding periportal biliary fibrosis. Whereas the extent of such biliary fibrosis is a negative predictor of long-term transplant-free survival, the cellular phenotypes involved in the fibrosis are not well established. Using a rhesus rotavirus-induced mouse model of BA, we demonstrate significant expansion of a cell population expressing the putative stem/progenitor cell marker, PROMININ-1 (PROM1), adjacent to ductular reactions within regions of periportal fibrosis. PROM1positive (pos) cells express Collagen-1α1. Subsets of PROM1pos cells coexpress progenitor cell marker CD49f, epithelial marker E-CADHERIN, biliary marker CYTOKERATIN-19, and mesenchymal markers VIMENTIN and alpha-SMOOTH MUSCLE ACTIN (αSMA). Expansion of the PROM1pos cell population is associated with activation of Fibroblast Growth Factor (FGF) and Transforming Growth Factor-beta (TGFβ) signaling. In vitro cotreatment of PROM1-expressing Mat1a-/- hepatic progenitor cells with recombinant human FGF10 and TGFβ1 promotes morphologic transformation toward a myofibroblastic cell phenotype with increased expression of myofibroblastic genes Collagen-1α1, Fibronectin, and α-Sma. Infants with BA demonstrate similar expansion of periportal PROM1pos cells with activated Mothers Against Decapentaplegic Homolog 3 (SMAD3) signaling in association with increased hepatic expression of FGF10, FGFR1, and FGFR2 as well as mesenchymal genes SLUG and SNAIL. Infants with perinatal subtype of BA have higher tissue levels of PROM1 expression than those with embryonic subtype. CONCLUSION Expansion of collagen-producing PROM1pos cells within regions of periportal fibrosis is associated with activated FGF and TGFβ pathways in both experimental and human BA. PROM1pos cells may therefore play an important role in the biliary fibrosis of BA.
Collapse
Affiliation(s)
- Nirmala Mavila
- Developmental Biology, Regenerative Medicine and Stem Cell Program, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Granier CJ, Wang W, Tsang T, Steward R, Sabaawy HE, Bhaumik M, Rabson AB. Conditional inactivation of PDCD2 induces p53 activation and cell cycle arrest. Biol Open 2014; 3:821-31. [PMID: 25150276 PMCID: PMC4163659 DOI: 10.1242/bio.20148326] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
PDCD2 (programmed cell death domain 2) is a highly conserved, zinc finger MYND domain-containing protein essential for normal development in the fly, zebrafish and mouse. The molecular functions and cellular activities of PDCD2 remain unclear. In order to better understand the functions of PDCD2 in mammalian development, we have examined PDCD2 activity in mouse blastocyst embryos, as well as in mouse embryonic stem cells (ESCs) and embryonic fibroblasts (MEFs). We have studied mice bearing a targeted PDCD2 locus functioning as a null allele through a splicing gene trap, or as a conditional knockout, by deletion of exon2 containing the MYND domain. Tamoxifen-induced knockout of PDCD2 in MEFs, as well as in ESCs, leads to defects in progression from the G1 to the S phase of cell cycle, associated with increased levels of p53 protein and p53 target genes. G1 prolongation in ESCs was not associated with induction of differentiation. Loss of entry into S phase of the cell cycle and marked induction of nuclear p53 were also observed in PDCD2 knockout blastocysts. These results demonstrate a unique role for PDCD2 in regulating the cell cycle and p53 activation during early embryonic development of the mouse.
Collapse
Affiliation(s)
- Celine J Granier
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Wei Wang
- Sequencing and Microarray Core Facility, Lewis-Sigler Institute for Integrative Genetics, Princeton University, Princeton, NJ 08854, USA
| | - Tiffany Tsang
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Ruth Steward
- Waksman Institute and Department of Molecular Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Hatem E Sabaawy
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08903, USA
| | - Mantu Bhaumik
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08903, USA
| | - Arnold B Rabson
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08903, USA
| |
Collapse
|
231
|
Sakao-Suzuki M, Kawasaki H, Akamatsu T, Meguro S, Miyajima H, Iwashita T, Tsutsui Y, Inoue N, Kosugi I. Aberrant fetal macrophage/microglial reactions to cytomegalovirus infection. Ann Clin Transl Neurol 2014; 1:570-88. [PMID: 25356429 PMCID: PMC4184560 DOI: 10.1002/acn3.88] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 07/07/2014] [Indexed: 12/20/2022] Open
Abstract
Objective Congenital cytomegalovirus (CMV) infection is the leading viral cause of neurodevelopmental disorders in humans, with the most severe and permanent sequelae being those affecting the cerebrum. As the fetal immune reactions to congenital CMV infection in the brain and their effects on cerebral development remain elusive, our aim was to investigate primitive innate immunity to CMV infection and its effects on cerebral corticogenesis in a mouse model for congenital CMV infection using a precise intraplacental inoculation method. Methods At 13.5 embryonic days (E13.5), pregnant C57BL/6 mice were intraplacentally infected with murine CMV (MCMV). Placentas and fetal organs were collected at 1, 3, and 5 days postinfection and analyzed. Results MCMV antigens were found frequently in perivascular macrophages, and subsequently in neural stem/progenitor cells (NSPCs). With increased expression of inducible nitric oxide synthase and proinflammatory cytokines, activated macrophages infiltrated into the infectious foci. In addition to the infected area, the numbers of both meningeal macrophages and parenchymal microglia increased even in the uninfected areas of MCMV-infected brain due to recruitment of their precursors from other sites. A bromodeoxyuridine (BrdU) incorporation experiment demonstrated that MCMV infection globally disrupted the self-renewal of NSPCs. Furthermore, BrdU-labeled neurons, particularly Brn2+ neurons of upper layers II/III in the cortical plate, decreased in number significantly in the MCMV-infected E18.5 cerebrum. Interpretation Brain macrophages are crucial for innate immunity during MCMV infection in the fetal brain, while their aberrant recruitment and activation may adversely impact on the stemness of NSPCs, resulting in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Makiko Sakao-Suzuki
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine Hamamatsu, Japan ; Department of Neurology, Hamamatsu University School of Medicine Hamamatsu, Japan
| | - Hideya Kawasaki
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine Hamamatsu, Japan
| | - Taisuke Akamatsu
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine Hamamatsu, Japan ; Department of Respiratory Medicine, Hamamatsu University School of Medicine Hamamatsu, Japan
| | - Shiori Meguro
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine Hamamatsu, Japan
| | - Hiroaki Miyajima
- Department of Neurology, Hamamatsu University School of Medicine Hamamatsu, Japan
| | - Toshihide Iwashita
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine Hamamatsu, Japan
| | | | - Naoki Inoue
- Department of Microbiology and Immunology, Gifu Pharmaceutical University Gifu, Japan
| | - Isao Kosugi
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine Hamamatsu, Japan
| |
Collapse
|
232
|
Lai YJ, Li MY, Yang CY, Huang KH, Tsai JC, Wang TW. TRIP6 regulates neural stem cell maintenance in the postnatal mammalian subventricular zone. Dev Dyn 2014; 243:1130-42. [DOI: 10.1002/dvdy.24161] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 06/25/2014] [Accepted: 06/25/2014] [Indexed: 11/06/2022] Open
Affiliation(s)
- Yun-Ju Lai
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Ming-Yang Li
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Cheng-Yao Yang
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Kao-Hua Huang
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Jui-Cheng Tsai
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Tsu-Wei Wang
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
- Brain Research Center; National Yang-Ming University; Taipei Taiwan
| |
Collapse
|
233
|
Abstract
Blood is renewed throughout the entire life. The stem cells of the blood, called hematopoietic stem cells (HSCs), are responsible for maintaining a supply of all types of fresh blood cells. In contrast to other stem cells, the clinical application of these cells is well established and HSC transplantation is an established life-saving therapy for patients suffering from haematological disorders. Despite their efficient functionality throughout life in vivo, controlling HSC behaviour in vitro (including their proliferation and differentiation) is still a major task that has not been resolved with standard cell culture systems. Targeted HSC multiplication in vitro could be beneficial for many patients, because HSC supply is limited. The biology of these cells and their natural microenvironment - their niche - remain a matter of ongoing research. In recent years, evidence has come to light that HSCs are susceptible to physical stimuli. This makes the regulation of HSCs by engineering physical parameters a promising approach for the targeted manipulation of these cells for clinical applications. Nevertheless, the biophysical regulation of these cells is still poorly understood. This review sheds light on the role of biophysical parameters in HSC biology and outlines which knowledge on biophysical regulation identified in other cell types could be applied to HSCs.
Collapse
Affiliation(s)
- C Lee-Thedieck
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany.
| | | |
Collapse
|
234
|
Similar morphological and molecular signatures shared by female and male germline stem cells. Sci Rep 2014; 4:5580. [PMID: 24993338 PMCID: PMC4082104 DOI: 10.1038/srep05580] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 06/18/2014] [Indexed: 11/30/2022] Open
Abstract
The existence of mammalian female germline stem cells (FGSCs) indicates that mammalian ovaries possess germline stem cells analogous to testis, and continue to produce gametes postnatally, which provides new insights into female fertility. In this study, we compared the morphological and molecular characteristics between FGSCs and spermatogonial stem cells (SSCs) by analysis of morphology, immunofluorescence, alkaline phosphatase activity assay, reverse transcription polymerase chain reaction (RT-PCR) and microarray hybridization. The results demonstrated that the morphology and growth patterns of FGSCs are similar to those of SSCs. Microarray analysis of global gene expression profiles of FGSCs and SSCs showed similar signatures in the transcriptome level. A list of 853 co-highly expressed genes (CEG) in female and male germline stem cells may be responsible for the morphological and molecular similarity. We constructed a continuous network of the CEG based on I2D protein-protein interaction database by breadth first search. From the network, we could observe the interactions of the CEG may be responsible for maintaining the properties of germline stem cells. This study was the first attempt to compare morphological and molecular characteristics between FGSCs and SSCs. These findings would provide some clues for further research on mammalian FGSCs.
Collapse
|
235
|
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
|
236
|
Wang H, Zhang Q, Fang X. Transcriptomics and proteomics in stem cell research. Front Med 2014; 8:433-44. [PMID: 24972645 DOI: 10.1007/s11684-014-0336-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 03/14/2014] [Indexed: 12/20/2022]
Abstract
Stem cells are capable of self-renewal and differentiation, and the processes regulating these events are among the most comprehensively investigated topics in life sciences. In particular, the molecular mechanisms of the self-renewal, proliferation, and differentiation of stem cells have been extensively examined. Multi-omics integrative analysis, such as transcriptomics combined with proteomics, is one of the most promising approaches to the systemic investigation of stem cell biology. We reviewed the available information on stem cells by examining published results using transcriptomic and proteomic characterization of the different stem cell processes. Comprehensive understanding of these important processes can only be achieved using a systemic methodology, and employing such method will strengthen the study on stem cell biology and promote the clinical applications of stem cells.
Collapse
Affiliation(s)
- Hai Wang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | | | | |
Collapse
|
237
|
Kim MJ, Kim MH, Kim SA, Chang JS. Age-related Deterioration of Hematopoietic Stem Cells. Int J Stem Cells 2014; 1:55-63. [PMID: 24855509 DOI: 10.15283/ijsc.2008.1.1.55] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2008] [Indexed: 01/12/2023] Open
Abstract
Aging is the process of system deterioration over time in the whole body. Stem cells are self-renewing and therefore have been considered exempt from the aging process. Earlier studies by Hayflick showed that there is an intrinsic limit to the number of divisions that mammalian somatic cells can undergo, and cycling kinetics and ontogeny-related studies strongly suggest that even the most primitive stem cell functions exhibit a certain degree of aging. Despite these findings, studies on the effects of aging on stem cell functions are inconclusive. Here we review the age-related properties of hematopoietic stem cells in terms of intrinsic and extrinsic alterations, proliferative potential, signaling molecules, telomere and telomerase, senescence and cancer issues, regenerative potential and other indications of stem cell aging are discussed in detail.
Collapse
Affiliation(s)
- Mi Jung Kim
- Department of Laboratory Medicine, University of Ulsan College of Medicine ; Cell and Molecular Biology Laboratory, Asan Institute for Life Sciences, AMC
| | - Min Hwan Kim
- Cell and Molecular Biology Laboratory, Asan Institute for Life Sciences, AMC
| | - Seung Ah Kim
- Cell and Molecular Biology Laboratory, Asan Institute for Life Sciences, AMC
| | - Jae Suk Chang
- Department of Orthopedic Surgery, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
238
|
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
|
239
|
Clark BS, Blackshaw S. Long non-coding RNA-dependent transcriptional regulation in neuronal development and disease. Front Genet 2014; 5:164. [PMID: 24936207 PMCID: PMC4047558 DOI: 10.3389/fgene.2014.00164] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 05/18/2014] [Indexed: 01/17/2023] Open
Abstract
Comprehensive analysis of the mammalian transcriptome has revealed that long non-coding RNAs (lncRNAs) may make up a large fraction of cellular transcripts. Recent years have seen a surge of studies aimed at functionally characterizing the role of lncRNAs in development and disease. In this review, we discuss new findings implicating lncRNAs in controlling development of the central nervous system (CNS). The evolution of the higher vertebrate brain has been accompanied by an increase in the levels and complexities of lncRNAs expressed within the developing nervous system. Although a limited number of CNS-expressed lncRNAs are now known to modulate the activity of proteins important for neuronal differentiation, the function of the vast majority of neuronal-expressed lncRNAs is still unknown. Topics of intense current interest include the mechanism by which CNS-expressed lncRNAs might function in epigenetic and transcriptional regulation during neuronal development, and how gain and loss of function of individual lncRNAs contribute to neurological diseases.
Collapse
Affiliation(s)
- Brian S Clark
- Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Seth Blackshaw
- Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Department of Ophthalmology, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Department of Neurology, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Center for High-Throughput Biology, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Institute for Cell Engineering, Johns Hopkins University School of Medicine Baltimore, MD, USA
| |
Collapse
|
240
|
Flach K, Ramminger E, Hilbrich I, Arsalan-Werner A, Albrecht F, Herrmann L, Goedert M, Arendt T, Holzer M. Axotrophin/MARCH7 acts as an E3 ubiquitin ligase and ubiquitinates tau protein in vitro impairing microtubule binding. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1527-38. [PMID: 24905733 PMCID: PMC4311138 DOI: 10.1016/j.bbadis.2014.05.029] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 05/05/2014] [Accepted: 05/28/2014] [Indexed: 12/11/2022]
Abstract
Tau is the major microtubule-associated protein in neurons involved in microtubule stabilization in the axonal compartment. Changes in tau gene expression, alternative splicing and posttranslational modification regulate tau function and in tauopathies can result in tau mislocalization and dysfunction, causing tau aggregation and cell death. To uncover proteins involved in the development of tauopathies, a yeast two-hybrid system was used to screen for tau-interacting proteins. We show that axotrophin/MARCH7, a RING-variant domain containing protein with similarity to E3 ubiquitin ligases interacts with tau. We defined the tau binding domain to amino acids 552–682 of axotrophin comprising the RING-variant domain. Co-immunoprecipitation and co-localization confirmed the specificity of the interaction. Intracellular localization of axotrophin is determined by an N-terminal nuclear targeting signal and a C-terminal nuclear export signal. In AD brain nuclear localization is lost and axotrophin is rather associated with neurofibrillary tangles. We find here that tau becomes mono-ubiquitinated by recombinant tau-interacting RING-variant domain, which diminishes its microtubule-binding. In vitro ubiquitination of four-repeat tau results in incorporation of up to four ubiquitin molecules compared to two molecules in three-repeat tau. In summary, we present a novel tau modification occurring preferentially on 4-repeat tau protein which modifies microtubule-binding and may impact on the pathogenesis of tauopathies. We search for tau-interacting proteins using a cytotrap yeast two-hybrid assay. MARCH7 was identified as a tau-binding protein and confirmed by several methods. Recombinant MARCH7 Ring-variant domain uses Ubc5 for E3 self-ubiquitinating activity. MARCH7 Ring-variant domain mono-ubiquitinates tau protein at multiple sites including the microtubule-binding domain. Mono-ubiquitination of tau protein diminishes its microtubule-binding.
Collapse
Affiliation(s)
- Katharina Flach
- Paul Flechsig Institute of Brain Research, Department of Molecular and Cellular Mechanisms of Neurodegeneration, University of Leipzig, 04109 Leipzig, Germany
| | - Ellen Ramminger
- Paul Flechsig Institute of Brain Research, Department of Molecular and Cellular Mechanisms of Neurodegeneration, University of Leipzig, 04109 Leipzig, Germany
| | - Isabel Hilbrich
- Paul Flechsig Institute of Brain Research, Department of Molecular and Cellular Mechanisms of Neurodegeneration, University of Leipzig, 04109 Leipzig, Germany
| | - Annika Arsalan-Werner
- Paul Flechsig Institute of Brain Research, Department of Molecular and Cellular Mechanisms of Neurodegeneration, University of Leipzig, 04109 Leipzig, Germany
| | - Franziska Albrecht
- Paul Flechsig Institute of Brain Research, Department of Molecular and Cellular Mechanisms of Neurodegeneration, University of Leipzig, 04109 Leipzig, Germany
| | - Lydia Herrmann
- Paul Flechsig Institute of Brain Research, Department of Molecular and Cellular Mechanisms of Neurodegeneration, University of Leipzig, 04109 Leipzig, Germany
| | - Michel Goedert
- MRC, Laboratory of Molecular Biology, Neurobiology Division, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Thomas Arendt
- Paul Flechsig Institute of Brain Research, Department of Molecular and Cellular Mechanisms of Neurodegeneration, University of Leipzig, 04109 Leipzig, Germany
| | - Max Holzer
- Paul Flechsig Institute of Brain Research, Department of Molecular and Cellular Mechanisms of Neurodegeneration, University of Leipzig, 04109 Leipzig, Germany.
| |
Collapse
|
241
|
Dingal PCDP, Discher DE. Combining insoluble and soluble factors to steer stem cell fate. NATURE MATERIALS 2014; 13:532-7. [PMID: 24845982 DOI: 10.1038/nmat3997] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Affiliation(s)
- P C Dave P Dingal
- Biophysical Engineering Laboratory in Chemical and Biomolecular Engineering, Philadelphia, Pennsylvania 19104, USA
| | - Dennis E Discher
- 1] Biophysical Engineering Laboratory in Chemical and Biomolecular Engineering, Philadelphia, Pennsylvania 19104, USA [2] Physics, and Cell and Molecular Biology Graduate Groups, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
242
|
Urine as a source of stem cells. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2014; 129:19-32. [PMID: 23038280 DOI: 10.1007/10_2012_157] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Traditionally, clinicians and researchers have relied on a skin biopsy or blood extraction as relatively accessible supplies for in vitro cell expansion and biological studies. Perhaps surprisingly, limited attention has been given to a totally noninvasive source, urine, which eliminates the discomfort associated with other procedures. This may arise from the perception that urine is merely a body waste. Yet, the analysis of urine is a longstanding fundamental test for diagnostic purposes and nowadays there is growing interest in using urine for detecting biomarkers. In addition, recent work including ours reinforces the idea that urine contains a variety of viable cell types with relevant applications. In this review, we describe those cell types and their potential uses.
Collapse
|
243
|
Abstract
The Hippo signaling pathway, consisting of a highly conserved kinase cascade (MST and Lats) and downstream transcription coactivators (YAP and TAZ), plays a key role in tissue homeostasis and organ size control by regulating tissue-specific stem cells. Moreover, this pathway plays a prominent role in tissue repair and regeneration. Dysregulation of the Hippo pathway is associated with cancer development. Recent studies have revealed a complex network of upstream inputs, including cell density, mechanical sensation, and G-protein-coupled receptor (GPCR) signaling, that modulate Hippo pathway activity. This review focuses on the role of the Hippo pathway in stem cell biology and its potential implications in tissue homeostasis and cancer.
Collapse
Affiliation(s)
- Jung-Soon Mo
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego La Jolla, CA, USA
| | - Hyun Woo Park
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego La Jolla, CA, USA
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego La Jolla, CA, USA
| |
Collapse
|
244
|
Sun GY, Dong LY, An W. Involvement of hepatic stimulator substance in the regulation of hepatoblast maturation into hepatocytes in vitro. Stem Cells Dev 2014; 23:1675-87. [PMID: 24640968 DOI: 10.1089/scd.2013.0468] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hepatic stimulator substance (HSS), also known as augmenter of liver regeneration (ALR), acts as a hepatotrophic growth factor to promote liver regeneration after liver damage or partial hepatectomy. However, the expression and function of HSS during liver development in mammals remain largely unknown. In this work, the hepatoblasts were isolated from mice at embryonic day 13.5 (E13.5), and HSS expression and its role during hepatoblast maturation were investigated. The results showed that HSS expression was enhanced in the hepatoblasts compared with mouse primary hepatocytes. HSS expression (23 kDa) was significantly decreased if the hepatoblast maturation was induced by a combination of oncostatin M (OSM), dexamethasone (DEX), and hepatocyte growth factor (HGF). We also found that knockdown of HSS expression (mainly 23-kDa isoform) by siRNA promoted hepatoblast maturation and also activated the signal transducer and activator of transcription 3 (STAT3) phosphorylation levels. However, if STAT3 activity was blocked by a small-molecule inhibitor Stattic, then hepatocyte maturation could be abolished, suggesting that STAT3 was most likely a potential molecule responsible for HSS signaling. In summary, our results demonstrated for the first time that HSS might be an active factor participating in the regulation of liver development and hepatocyte maturation.
Collapse
Affiliation(s)
- Guang-Yong Sun
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regeneration Regulation, Capital Medical University , Beijing, China
| | | | | |
Collapse
|
245
|
Filioli Uranio M, Dell'Aquila ME, Caira M, Guaricci AC, Ventura M, Catacchio CR, Martino NA, Valentini L. Characterization and in vitro differentiation potency of early-passage canine amnion- and umbilical cord-derived mesenchymal stem cells as related to gestational age. Mol Reprod Dev 2014; 81:539-51. [PMID: 24659564 DOI: 10.1002/mrd.22322] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 03/15/2014] [Indexed: 12/20/2022]
Abstract
Fetal adnexa are a non-controversial source of mesenchymal stem cells (MSCs) that have high plasticity, a high proliferation rate, and the ability to differentiate towards multiple lineages. MSC populations have been characterized for their stemness and differentiation capabilities; more recent work has focused on MSC selection and on establishing predictable elements to discriminate the cells with the most potential for regenerative medicine. In this study, we cytogenetically and molecularly characterized and followed the in vitro proliferation and differentiation potential of early-passage canine amniotic membrane MSCs (AM-MSCs) and umbilical cord matrix MSCs (UCM-MSCs) isolated from fetuses at early (35-40 days) and late (45-55 days) gestational ages. We found that cells from both fetal gestational ages showed similar features. In all examined cell lines, the morphology of proliferating cells typically appeared fibroblast-like. Population doublings, passaged up to 10 times, increased significantly with passage number. In both cell types, cell viability and chromosomal number and structure were not affected by gestational age at early passages. Passage-3 AM- and UCM-MSCs from both gestational phases also expressed embryonic (POU5F1) and mesenchymal (CD29, CD44) stemness markers, whereas hematopoietic and histocompatibility markers were never found in any sample. Passage-3 cell populations of each cell type were also multipotential as they could differentiate into neurocytes and osteocytes, based on cell morphology, specific stains, and molecular analysis. These results indicated that MSCs retrieved from the UCM and AM in the early and late fetal phases of gestation could be used for canine regenerative medicine.
Collapse
Affiliation(s)
- Manuel Filioli Uranio
- Veterinary Clinics and Animal Productions Section, Department for Emergency and Organ Transplantation, University of Bari Aldo Moro, Valenzano, Italy
| | | | | | | | | | | | | | | |
Collapse
|
246
|
Chung KM, Kolling IV FW, Gajdosik MD, Burger S, Russell AC, Nelson CE. Single cell analysis reveals the stochastic phase of reprogramming to pluripotency is an ordered probabilistic process. PLoS One 2014; 9:e95304. [PMID: 24743916 PMCID: PMC3990627 DOI: 10.1371/journal.pone.0095304] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 03/26/2014] [Indexed: 12/27/2022] Open
Abstract
Despite years of research, the reprogramming of human somatic cells to pluripotency remains a slow, inefficient process, and a detailed mechanistic understanding of reprogramming remains elusive. Current models suggest reprogramming to pluripotency occurs in two-phases: a prolonged stochastic phase followed by a rapid deterministic phase. In this paradigm, the early stochastic phase is marked by the random and gradual expression of pluripotency genes and is thought to be a major rate-limiting step in the successful generation of induced Pluripotent Stem Cells (iPSCs). Recent evidence suggests that the epigenetic landscape of the somatic cell is gradually reset during a period known as the stochastic phase, but it is known neither how this occurs nor what rate-limiting steps control progress through the stochastic phase. A precise understanding of gene expression dynamics in the stochastic phase is required in order to answer these questions. Moreover, a precise model of this complex process will enable the measurement and mechanistic dissection of treatments that enhance the rate or efficiency of reprogramming to pluripotency. Here we use single-cell transcript profiling, FACS and mathematical modeling to show that the stochastic phase is an ordered probabilistic process with independent gene-specific dynamics. We also show that partially reprogrammed cells infected with OSKM follow two trajectories: a productive trajectory toward increasingly ESC-like expression profiles or an alternative trajectory leading away from both the fibroblast and ESC state. These two pathways are distinguished by the coordinated expression of a small group of chromatin modifiers in the productive trajectory, supporting the notion that chromatin remodeling is essential for successful reprogramming. These are the first results to show that the stochastic phase of reprogramming in human fibroblasts is an ordered, probabilistic process with gene-specific dynamics and to provide a precise mathematical framework describing the dynamics of pluripotency gene expression during reprogramming by OSKM.
Collapse
Affiliation(s)
- Kyung-Min Chung
- University of Connecticut Department of Molecular and Cell biology, Storrs, Connecticut, United States of America
| | - Frederick W. Kolling IV
- University of Connecticut Department of Molecular and Cell biology, Storrs, Connecticut, United States of America
| | - Matthew D. Gajdosik
- University of Connecticut Department of Molecular and Cell biology, Storrs, Connecticut, United States of America
| | - Steven Burger
- University of Connecticut Department of Molecular and Cell biology, Storrs, Connecticut, United States of America
| | - Alexander C. Russell
- University of Connecticut Department of Computer Science and Engineering, Storrs, Connecticut, United States of America
| | - Craig E. Nelson
- University of Connecticut Department of Molecular and Cell biology, Storrs, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
247
|
|
248
|
Abstract
Despite advances in understanding pluripotency through traditional cell biology and gene expression profiling, the signaling networks responsible for maintenance of pluripotency and lineage-specific differentiation are poorly defined. To aid in an improved understanding of these networks at the systems level, we present procedures for the combined analysis of the total proteome and total phosphoproteome (termed (phospho)proteome) from human embryonic stem cells (hESCs), human induced pluripotent stem cells (hiPSCs), and their differentiated derivatives. Because there has been considerable heterogeneity in the literature on the culture of pluripotent cells, we first briefly describe our feeder-free cell culture protocol. The focus, however, is on procedures necessary to generate large-scale (phospho)proteomic data from the cells. Human cells are described here, but the (phospho)proteomic procedures are broadly applicable. Detailed procedures are given for lysis of the cells, protein sample preparation and digestion, multidimensional liquid chromatography, analysis by tandem mass spectrometry, and database searches for peptide/protein identification (ID). We summarize additional data analysis procedures, the subject of ongoing efforts.
Collapse
|
249
|
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: 183] [Impact Index Per Article: 16.6] [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
|
250
|
Minakhina S, Changela N, Steward R. Zfrp8/PDCD2 is required in ovarian stem cells and interacts with the piRNA pathway machinery. Development 2014; 141:259-68. [PMID: 24381196 DOI: 10.1242/dev.101410] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The maintenance of stem cells is central to generating diverse cell populations in many tissues throughout the life of an animal. Elucidating the mechanisms involved in how stem cells are formed and maintained is crucial to understanding both normal developmental processes and the growth of many cancers. Previously, we showed that Zfrp8/PDCD2 is essential for the maintenance of Drosophila hematopoietic stem cells. Here, we show that Zfrp8/PDCD2 is also required in both germline and follicle stem cells in the Drosophila ovary. Expression of human PDCD2 fully rescues the Zfrp8 phenotype, underlining the functional conservation of Zfrp8/PDCD2. The piRNA pathway is essential in early oogenesis, and we find that nuclear localization of Zfrp8 in germline stem cells and their offspring is regulated by some piRNA pathway genes. We also show that Zfrp8 forms a complex with the piRNA pathway protein Maelstrom and controls the accumulation of Maelstrom in the nuage. Furthermore, Zfrp8 regulates the activity of specific transposable elements also controlled by Maelstrom and Piwi. Our results suggest that Zfrp8/PDCD2 is not an integral member of the piRNA pathway, but has an overlapping function, possibly competing with Maelstrom and Piwi.
Collapse
Affiliation(s)
- Svetlana Minakhina
- Rutgers University, Department of Molecular Biology, Waksman Institute, Cancer Institute of New Jersey, 190 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | | | | |
Collapse
|